Network


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

Hotspot


Dive into the research topics where Gabriel A. Kwong is active.

Publication


Featured researches published by Gabriel A. Kwong.


Nature Biotechnology | 2008

Integrated barcode chips for rapid, multiplexed analysis of proteins in microliter quantities of blood

Rong Fan; Ophir Vermesh; Alok Srivastava; Brian K.H. Yen; Lidong Qin; Habib Ahmad; Gabriel A. Kwong; Chao Chao Liu; Juliane Gould; Leroy Hood; James R. Heath

As the tissue that contains the largest representation of the human proteome, blood is the most important fluid for clinical diagnostics. However, although changes of plasma protein profiles reflect physiological or pathological conditions associated with many human diseases, only a handful of plasma proteins are routinely used in clinical tests. Reasons for this include the intrinsic complexity of the plasma proteome, the heterogeneity of human diseases and the rapid degradation of proteins in sampled blood. We report an integrated microfluidic system, the integrated blood barcode chip that can sensitively sample a large panel of protein biomarkers over broad concentration ranges and within 10 min of sample collection. It enables on-chip blood separation and rapid measurement of a panel of plasma proteins from quantities of whole blood as small as those obtained by a finger prick. Our device holds potential for inexpensive, noninvasive and informative clinical diagnoses, particularly in point-of-care settings.Blood comprises the largest version of the human proteome1. Changes of plasma protein profiles can reflect physiological or pathological conditions associated with many human diseases, making blood the most important fluid for clinical diagnostics2-4. Nevertheless, only a handful of plasma proteins are utilized in routine clinical tests. This is due to a host of reasons, including the intrinsic complexity of the plasma proteome1, the heterogeneity of human diseases and the fast kinetics associated with protein degradation in sampled blood5. Simple technologies that can sensitively sample large numbers of proteins over broad concentration ranges, from small amounts of blood, and within minutes of sample collection, would assist in solving these problems. Herein, we report on an integrated microfluidic system, called the Integrated Blood Barcode Chip (IBBC). It enables on-chip blood separation and the rapid measurement of a panel of plasma proteins from small quantities of blood samples including a fingerprick of whole blood. This platform holds potential for inexpensive, non-invasive, and informative clinical diagnoses, particularly, for point-of-care.


Nature Medicine | 2011

A clinical microchip for evaluation of single immune cells reveals high functional heterogeneity in phenotypically similar T cells.

Chao Ma; Rong Fan; Habib Ahmad; Qihui Shi; Begonya Comin-Anduix; Thinle Chodon; Richard C. Koya; Chao-Chao Liu; Gabriel A. Kwong; Caius G. Radu; Antoni Ribas; James R. Heath

Cellular immunity has an inherent high level of functional heterogeneity. Capturing the full spectrum of these functions requires analysis of large numbers of effector molecules from single cells. We report a microfluidic platform designed for highly multiplexed (more than ten proteins), reliable, sample-efficient (∼1 × 104 cells) and quantitative measurements of secreted proteins from single cells. We validated the platform by assessment of multiple inflammatory cytokines from lipopolysaccharide (LPS)-stimulated human macrophages and comparison to standard immunotechnologies. We applied the platform toward the ex vivo quantification of T cell polyfunctional diversity via the simultaneous measurement of a dozen effector molecules secreted from tumor antigen–specific cytotoxic T lymphocytes (CTLs) that were actively responding to tumor and compared against a cohort of healthy donor controls. We observed profound, yet focused, functional heterogeneity in active tumor antigen–specific CTLs, with the major functional phenotypes quantitatively identified. The platform represents a new and informative tool for immune monitoring and clinical assessment.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Point-of-care diagnostics for noncommunicable diseases using synthetic urinary biomarkers and paper microfluidics

Andrew D. Warren; Gabriel A. Kwong; David K. Wood; Kevin Y. Lin; Sangeeta N. Bhatia

Significance Noncommunicable diseases, including cardiovascular disease and cancer, are growing worldwide but are challenging to diagnose because biomarkers that can accurately detect them in patients are lacking. Here, we designed nanoscale agents that are administered to reveal the presence of diseased tissues by producing a biomarker in the urine that can be detected using paper strips similar to a home pregnancy test. Using mouse models, we show that we can detect diseases as diverse as solid cancer and blood clots using only a single injection of our diagnostic followed by urine analysis on paper. This platform does not require expensive instruments, invasive procedures, or trained medical personnel, and may allow low-cost diagnosis of diseases at the point of care in resource-limited settings. With noncommunicable diseases (NCDs) now constituting the majority of global mortality, there is a growing need for low-cost, noninvasive methods to diagnose and treat this class of diseases, especially in resource-limited settings. Molecular biomarkers combined with low-cost point-of-care assays constitute a potential solution for diagnosing NCDs, but the dearth of naturally occurring, predictive markers limits this approach. Here, we describe the design of exogenous agents that serve as synthetic biomarkers for NCDs by producing urinary signals that can be quantified by a companion paper test. These synthetic biomarkers are composed of nanoparticles conjugated to ligand-encoded reporters via protease-sensitive peptide substrates. Upon delivery, the nanoparticles passively target diseased sites, such as solid tumors or blood clots, where up-regulated proteases cleave the peptide substrates and release reporters that are cleared into urine. The reporters are engineered for detection by sandwich immunoassays, and we demonstrate their quantification directly from unmodified urine; furthermore, capture antibody specificity allows the probes to be multiplexed in vivo and quantified simultaneously by ELISA or paper lateral flow assay (LFA). We tailor synthetic biomarkers specific to colorectal cancer, a representative solid tumor, and thrombosis, a common cardiovascular disorder, and demonstrate urinary detection of these diseases in mouse models by paper diagnostic. Together, the LFA and injectable synthetic biomarkers, which could be tailored for multiple diseases, form a generalized diagnostic platform for NCDs that can be applied in almost any setting without expensive equipment or trained medical personnel.


ACS Nano | 2013

Nanoparticles that sense thrombin activity as synthetic urinary biomarkers of thrombosis.

Kevin Y. Lin; Gabriel A. Kwong; Andrew D. Warren; David K. Wood; Sangeeta N. Bhatia

Thrombin is a serine protease and regulator of hemostasis that plays a critical role in the formation of obstructive blood clots, or thrombosis, that is a life-threatening condition associated with numerous diseases such as atherosclerosis and stroke. To detect thrombi in living animals, we design and conjugate thrombin-sensitive peptide substrates to the surface of nanoparticles. Following intravenous infusion, these “synthetic biomarkers” survey the host vasculature for coagulation and, in response to substrate cleavage by thrombin, release ligand-encoded reporters into the host urine. To detect the urinary reporters, we develop a companion 96-well immunoassay that utilizes antibodies to bind specifically to the ligands, thus capturing the reporters for quantification. Using a thromboplastin-induced mouse model of pulmonary embolism, we show that urinary biomarker levels differentiate between healthy and thrombotic states and correlate closely with the aggregate burden of clots formed in the lungs. Our results demonstrate that synthetic biomarkers can be engineered to sense vascular diseases remotely from the urine and may allow applications in point-of-care diagnostics.


Journal of the American Chemical Society | 2011

Iterative in Situ Click Chemistry Assembles a Branched Capture Agent and Allosteric Inhibitor for Akt1

Steven W. Millward; Ryan K. Henning; Gabriel A. Kwong; Suresh M. Pitram; Heather D. Agnew; Kaycie M. Deyle; Arundhati Nag; Jason E. Hein; Su Seong Lee; Jaehong Lim; Jessica A. Pfeilsticker; K. Barry Sharpless; James R. Heath

We describe the use of iterative in situ click chemistry to design an Akt-specific branched peptide triligand that is a drop-in replacement for monoclonal antibodies in multiple biochemical assays. Each peptide module in the branched structure makes unique contributions to affinity and/or specificity resulting in a 200 nM affinity ligand that efficiently immunoprecipitates Akt from cancer cell lysates and labels Akt in fixed cells. Our use of a small molecule to preinhibit Akt prior to screening resulted in low micromolar inhibitory potency and an allosteric mode of inhibition, which is evidenced through a series of competitive enzyme kinetic assays. To demonstrate the efficiency and selectivity of the protein-templated in situ click reaction, we developed a novel QPCR-based methodology that enabled a quantitative assessment of its yield. These results point to the potential for iterative in situ click chemistry to generate potent, synthetically accessible antibody replacements with novel inhibitory properties.


Angewandte Chemie | 2011

High‐Density, Multiplexed Patterning of Cells at Single‐Cell Resolution for Tissue Engineering and Other Applications

Udi Vermesh; Ophir Vermesh; Jun Wang; Gabriel A. Kwong; Chao Ma; Kiwook Hwang; James R. Heath

Surface chemistry meets tissue engineering: A novel surface-patterning approach for creating arrays of DNA squares is combined with a unique method for DNA-encoding of cells to construct dense arrays of distinct single cells. The cell patterns can be transferred from the substrate surface into thin hydrogel films, and these layers can be stacked to form 3D tissue constructs.


Journal of the American Chemical Society | 2014

Disease detection by ultrasensitive quantification of microdosed synthetic urinary biomarkers.

Andrew D. Warren; Shonda T. Gaylord; Kevin C. Ngan; Milena Dumont Milutinovic; Gabriel A. Kwong; Sangeeta N. Bhatia; David R. Walt

The delivery of exogenous agents can enable noninvasive disease monitoring, but existing low-dose approaches require complex infrastructure. In this paper, we describe a microdose-scale injectable formulation of nanoparticles that interrogate the activity of thrombin, a key regulator of clotting, and produce urinary reporters of disease state. We establish a customized single molecule detection assay that enables urinary discrimination of thromboembolic disease in mice using doses of the nanoparticulate diagnostic agents that fall under regulatory guidelines for “microdosing.”


ACS Nano | 2014

Self-Titrating Anticoagulant Nanocomplexes That Restore Homeostatic Regulation of the Coagulation Cascade

Kevin Y. Lin; Justin H. Lo; Nikita Consul; Gabriel A. Kwong; Sangeeta N. Bhatia

Antithrombotic therapy is a critical portion of the treatment regime for a number of life-threatening conditions, including cardiovascular disease, stroke, and cancer; yet, proper clinical management of anticoagulation remains a challenge because existing agents increase the propensity for bleeding in patients. Here, we describe the development of a bioresponsive peptide–polysaccharide nanocomplex that utilizes a negative feedback mechanism to self-titrate the release of anticoagulant in response to varying levels of coagulation activity. This nanoscale self-titrating activatable therapeutic, or nanoSTAT, consists of a cationic thrombin-cleavable peptide and heparin, an anionic polysaccharide and widely used clinical anticoagulant. Under nonthrombotic conditions, nanoSTATs circulate inactively, neither releasing anticoagulant nor significantly prolonging bleeding time. However, in response to life-threatening pulmonary embolism, nanoSTATs locally release their drug payload and prevent thrombosis. This autonomous negative feedback regulator may improve antithrombotic therapy by increasing the therapeutic window and decreasing the bleeding risk of anticoagulants.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Mathematical framework for activity-based cancer biomarkers

Gabriel A. Kwong; Jaideep S. Dudani; Emmanuel Carrodeguas; Eric V. Mazumdar; Seyedeh M. Zekavat; Sangeeta N. Bhatia

Significance The discovery of cancer at an early stage improves treatment outcomes, yet cancer detection thresholds based on measuring the abundance of biomarkers produced by small tumors are biologically limited. Here we develop a mathematical framework to explore the use of activity-based biomarkers for early cancer detection. In contrast to abundance-based biomarkers, activity-based biomarkers rely on the catalytic activity of enzymes to amplify cancer-derived signals and allow detection of small tumors. Using a class of activity-based biomarkers called synthetic biomarkers, we comprehensively explore how detection sensitivities depend on probe design, enzymatic activity, and organ physiology, and how they may be precisely tuned to reveal the presence of small tumors in humans. Advances in nanomedicine are providing sophisticated functions to precisely control the behavior of nanoscale drugs and diagnostics. Strategies that coopt protease activity as molecular triggers are increasingly important in nanoparticle design, yet the pharmacokinetics of these systems are challenging to understand without a quantitative framework to reveal nonintuitive associations. We describe a multicompartment mathematical model to predict strategies for ultrasensitive detection of cancer using synthetic biomarkers, a class of activity-based probes that amplify cancer-derived signals into urine as a noninvasive diagnostic. Using a model formulation made of a PEG core conjugated with protease-cleavable peptides, we explore a vast design space and identify guidelines for increasing sensitivity that depend on critical parameters such as enzyme kinetics, dosage, and probe stability. According to this model, synthetic biomarkers that circulate in stealth but then activate at sites of disease have the theoretical capacity to discriminate tumors as small as 5 mm in diameter—a threshold sensitivity that is otherwise challenging for medical imaging and blood biomarkers to achieve. This model may be adapted to describe the behavior of additional activity-based approaches to allow cross-platform comparisons, and to predict allometric scaling across species.


ACS Nano | 2015

Photoactivated Spatiotemporally-Responsive Nanosensors of in Vivo Protease Activity

Jaideep S. Dudani; Piyush K. Jain; Gabriel A. Kwong; Kelly R. Stevens; Sangeeta N. Bhatia

Proteases play diverse and important roles in physiology and disease, including influencing critical processes in development, immune responses, and malignancies. Both the abundance and activity of these enzymes are tightly regulated and highly contextual; thus, in order to elucidate their specific impact on disease progression, better tools are needed to precisely monitor in situ protease activity. Current strategies for detecting protease activity are focused on functionalizing synthetic peptide substrates with reporters that emit detection signals following peptide cleavage. However, these activity-based probes lack the capacity to be turned on at sites of interest and, therefore, are subject to off-target activation. Here we report a strategy that uses light to precisely control both the location and time of activity-based sensing. We develop photocaged activity-based sensors by conjugating photolabile molecules directly onto peptide substrates, thereby blocking protease cleavage by steric hindrance. At sites of disease, exposure to ultraviolet light unveils the nanosensors to allow proteases to cleave and release a reporter fragment that can be detected remotely. We apply this spatiotemporally controlled system to probe secreted protease activity in vitro and tumor protease activity in vivo. In vitro, we demonstrate the ability to dynamically and spatially measure metalloproteinase activity in a 3D model of colorectal cancer. In vivo, veiled nanosensors are selectively activated at the primary tumor site in colorectal cancer xenografts to capture the tumor microenvironment-enriched protease activity. The ability to remotely control activity-based sensors may offer a valuable complement to existing tools for measuring biological activity.

Collaboration


Dive into the Gabriel A. Kwong's collaboration.

Top Co-Authors

Avatar

Sangeeta N. Bhatia

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

James R. Heath

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Andrew D. Warren

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Caius G. Radu

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antoni Ribas

University of California

View shared research outputs
Top Co-Authors

Avatar

Chao Ma

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Geoffrey von Maltzahn

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Jaideep S. Dudani

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Owen N. Witte

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge