<<

MEET THE FACULTY CANDIDATES

Candidates will be present to meet with faculty recruiters on Wednesday, October 16, 2019 from 3:30pm – 5:30pm in Exhibit Hall DE. Admission to the Poster Forum (other than candidates below) are by a business card showing that the individual is a faculty

recruiter and a valid BMES Annual Meeting badge. AMR ASHRAF ABDEEN, PhD Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI . [email protected]

Research Overview: My interests lie at a unique intersection of biomaterials and mechanobiology, genomics and therapies. I aim to advance the analysis of biomaterial-cell interaction, fundamental mechanobiology and biomaterials-based therapeutics using novel genome methods in the biomaterials field. My doctoral work focused on the use of biomaterials to probe cell-matrix interaction – how cell matrix affects their differentiation and secretory profile for therapeutic purposes. My postdoctoral work focuses on the more translational usage of biomaterials for therapies. Here, I’ve worked on protein delivery for genome editing, had exposure to state-of the-art regenerative stem cell therapies (for in vivo implantation) and immunotherapies (such as CAR-T therapies). In addition, I now have extensive experience in CRISPR-based genome manipulation for fundamental studies as well as therapeutic purposes. This work involved learning more sophisticated, high-throughput molecular biology assays. One focus of my lab will be studying the more fundamental biomaterials-cell interactions using unbiased biological methods such as high throughput sequencing or functional screens, coupled with a library of biomaterials that covers a wide range of materials properties. Another focus will be the more translational aspect of therapeutics, with a focus on delivery techniques for both gene and cell therapies. Our more fundamental work will inform the design of biomaterials for translation.

Education: 2016 | Ph.D., and Engineering, University of Illinois at Urbana-Champaign 2010 | B.S., Materials Science and Engineering, German University in Cairo

Research/Work Experience: 2016 - Present | Postdoctoral Researcher Wisconsin Institute for Discovery, University of Wisconsin – Madison (Advisor: Krishanu Saha) 2011 - 2016 | Graduate Research Assistant Materials Science and Engineering, University of Illinois at Urbana-Champaign (Advisor: Kristopher Kilian)

Selected Publications: 1. G. Chen*, A. A. Abdeen*, Y. Wang*, P. Shahi, S. Robertson, R. Xie, M. Suzuki, B. Pattnaik, K. Saha, S. Gong (2019). " A biodegradable nanocapsule delivers a Cas9 ribonucleoprotein complex for in vivo genome editing", Nature (Accepted). 2. A. A. Abdeen*, J. Lee*, Y. Li, K. A. Kilian (2017). "Cytoskeletal priming of mesenchymal stem cells to a medicinal phenotype", Regenerative Engineering and Translational Medicine, 3(1) p5-14. 3. A. A. Abdeen, J. Lee, N. A. Bharadwaj, R. Ewoldt, K. A. Kilian (2016). "Temporal modulation of stem cell activity using magnetoactive hydrogels", Advanced Healthcare Materials, 5(19) p2536-2544. 4. J. Lee, A. A. Abdeen, K. L. Wycislo, T. M. Fan, K. A. Kilian (2016). "Interfacial geometry dictates cancer cell tumorigenicity", Nature MAterials, 15(8) p856. 5. A. A. Abdeen, J. B. Weiss, J. Lee, K. A. Kilian (2014). "Matrix composition and mechanics direct proangiogenic signaling from mesenchymal stem cells", Engineering A, 20(19-20) p2737-2745.

Awards/Honors: -Fellowship to attend GEM4 Mechanobiology of the Brain Workshop, Carnegie-Mellon University, Pittsburgh, PA (2015). -Racheff Teaching Fellowship, University of Illinois at Urbana-Champaign (2013). DANIEL ABEBAYEHU, PhD , University of Virginia, 415 Lane Road, Room 1225, Charlottesville, VA, 22908 [email protected]

Research Overview: My future goal is to become an independent investigator as a tenure-track professor at a major research university. As a professor, my research interests would entail exploring the intersection of innate immunity and to improve tissue repair and wound healing. In particular, I am interested in helping to unravel the complexity behind the immuno-stromal axis and determining avenues for intervention, as well as design immunomodulatory biomaterials that promote healing and repair. My academic and research training have provided me with a strong background in biomaterial fabrication, molecular biology, and immunology.

Education: B.S. Biomedical Engineering, 2011, University of Virginia PhD Biomedical Engineering, 2017, Virginia Commonwealth University

Research/Work Experience: 2019 – Present NRSA NHLBI Ruth L. Kirchstein Postdoc Fellow at UVA 2017 – 2019 Postdoc Fellow on T32 Cardiovascular Research Training Grant at UVA 2015 – 2017 Initiative for Maximizing Student Development (IMSD) PhD Fellow 2013 – 2015 Teaching Assistant, Department of Biomedical Engineering, Virginia Commonwealth University (Courses taught: EGRB101 Biomedical Engineering Practicuum I, EGRB102 Intro to Engineering, and EGRB303 Biotransport) 2011 – 2013 Graduate Research Assistant, Department of Biomedical Engineering, Virginia Commonwealth University 2010 NSF REU Summer Researcher, Department of Biomedical Engineering, University of Virginia 2008 – 2011 Undergraduate Research Assistant, Department of Biomedical Engineering, University of Virginia

Selected Publications: • Abebayehu D, Spence AJ, McClure MJ, Haque TT, Rivera K, and Ryan JJ (2019). “Polymer Scaffold Architecture is a Key Determinant in Mast Cell Inflammatory and Angiogenic Responses” Journal of Biomedical Materials Research Part A, 107(4): 884- 892. • Witherel CE, Abebayehu D, Barker TH, and Spiller KL (2019). “Macrophage and interactions in biomaterialmediated fibrosis.” Advanced Healthcare Materials, 8(4): e1801451. • Abebayehu D, Spence AJ, Caslin HL, Taruselli MT, Haque T, Kiwanuka K, Kolawole EM, Chumanevich A, Sell SA, Oskeritzian C, and Ryan JJ (2019). “Lactic acid suppresses inflammatory responses and enhances angiogenic effects of IgE-stimlated mast cells.” Cellular Immunology, pii: S0008-8749. • Caslin HL, Abebayehu D, Qayum AA, Haque TT, Taruselli MT, Paez PA, Pondicherry N, Barnstein BO, Hoeferlin LA, Chalfant CE, and Ryan JJ (2019). “Lactic acid inhibits LPS-induced mast cell function by limiting glycolysis and ATP availability.” Journal of Immunology, ji1801005. • Abebayehu D, Spence AJ, Boyan BD, Schwartz Z, Ryan JJ, and McClure MJ (2017). “Galectin-1 Promotes and M2 Macrophage Response to Polydioxanone Scaffolds.” Journal of Biomedical Materials Research Part A, 105(9): 2562-2571. • Abebayehu D, Ndaw VS, Spence AJ, Paez PA, Kolawole EM, Taruselli MT, Caslin HL, Chumanevich AP, Paranjape A, Baker B, Barnstein BO, Oskeritzian CA, and Ryan JJ (2017). “TGFβ1 Suppresses IL-33-induced Mast Cell Function.” Journal of Immunology, 199(3): 866-873. • Abebayehu D, Spence AJ, Abdul Qayum A, Taruselli MT, McLeod JJA, Caslin HL, Chumanveich AP, Montunrayo EM, Paranjape A, Baker B, Ndaw VS, Barnstein BO, Oskeritzian CA, Sell SA, and Ryan JJ (2016). “Lactic Acid Suppresses IL-33-mediated Mast Cell Inflammatory Responses via Hypoxia Inducible Factor (HIF)-1-dependent miR-155 Suppression” Journal of Immunology, 197(7): 2909-17.

Awards/Honors: F32 NRSA Ruth L. Kirchstein Postdoctoral Fellowship Travel award to Hilton Head Regenerative Medicine Workshop T32 Postdoc fellowship Cardiovascular Research Training Grant Initiative for Maximizing Student Development (IMSD) PhD Fellowship American Association of Immunologists Annual Conference Trainee Poster Award 1ST place award winner at the VCU Graduate Student Association Research Symposium Federation of American Societies for Experimental Biology/Maximizing Access to Research Careers Travel Award Winner ($1850) Finalist at the TERMIS-NA meeting poster competition in the Student and Young Investigator Section ALEXANDRE ALBANESE, PhD Institute for Medical Engineering and Science, MIT, E25-353, 77 Mass Ave, Cambridge, MA, 02139 [email protected] Research Overview: Organoid models provide a unique opportunity to study the complex cellular landscapes that underlie human biology. My research program will apply high-resolution fluorescence microscopy to analyze the spatial context of individual cells inside intact organoids to model patient-specific diseases. This research will combine my experience in 3D culture models, tissue clearing and biomaterial engineering to quantify the coordination of cellular networks inside human tissues. My PhD work with Prof. Warren Chan (University of Toronto) involved the development of a tumor-on-a-chip model to quantify nanoparticle accumulation and interactions inside the tumor microenvironment. My postdoctoral research continued to explore 3D culture models with Kwanghun Chung (MIT) where I developed a pipeline for high-dimensional analysis of intact human cerebral organoids using tissue clearing, light-sheet microscopy and algorithmic image analysis. Building on this work, my goal is to apply volumetric imaging of organoids to quantify three-dimensional cellular organization and responses in tissue models. This research will pursue three aims. (1) Development of a high-throughput organoid imaging pipeline. By correlating each cell's phenotype and context, it will be possible to infer cell populations from 2-3 strategic markers. This approach will be used to build a “flow cytometer” for organoids to enable rapid analysis. (2) Development of frameworks to correlate single-cell atlases with time-lapse data of organoid growth and maturation. The goal is to monitor how the evolution of anatomical features (e.g. ventricles) relate to cell phenotypes. (3) Generation of a 4D cell atlas capturing whole-organoid responses to location- specific stimuli (engineered using nanoparticles, bio-compatible scaffolds and perfusion-based culture systems). The goal is to characterize whole-tissue signal propagation at the cellular scale. My research will initially focus on human cerebral organoids to understand how complex cellular landscapes contribute to brain development. My research will eventually transition to patient-derived tumor organoids to evaluate the role of cellular organization in tumor growth and response to therapeutics.

Education: PhD, Biomedical Engineering (IBBME),2014, University of Toronto, Canada MSc, Microbiology & Immunology, 2006, McGill University, Canada BSc, Microbiology & Immunology, 2004, McGill University, Canada

Research/Work Experience: Postdoctoral Fellow, Cerebral organoid culture and volumetric imaging, MIT, IMES. Supervisor: Kwanghun Chung (2015- present) Postdoctoral Fellow, in vitro 3D cancer models to evaluate nanoparticles, MIT, IMES. Supervisor: Sangheeta Bhatia (2014- 2015) Research Technician, cell response to immuno-suppressive liposomes, Vasogen Inc. Toronto, Canada (2008) Associate Scientist, inflammatory response to purified proteins, GlaxoSmithKline Biologicals. Montreal, Canada (2006-2008)

Selected Publications: [1] Albanese A*, Swaney JM* [...]Chung K. 3D imaging and high-content analysis of intact human cerebral organoids. [Manuscript in preparation] [2] Yun DH, Park YG, Cho JH, Kamentsky L, Evans NB, Albanese A [...]Chung K. Ultrafast immunostaining of organ-scale tissues for scalable proteomic phenotyping. [in revision; pre-print available in bioRxiv] [3] Mano T*, Albanese A* [...]Chung K, Ueda HR. Whole-Brain Analysis of Cells and Circuits by Tissue Clearing and Light- Sheet Microscopy. J Neurosci. 2018 [4] Ku T*, Swaney J*, Park JY*, Albanese A [...] Chung K, Multiplexed and scalable super-resolution imaging of three- dimensional protein localization in size-adjustable tissues. Nat Biotechnol. 2016 [5] Albanese A*, Walkey CD*, Olsen J, Gao H, Emili A, and Chan WC. Secreted cellular proteins alter nanoparticle-cell interactions. ACS Nano. 2014 [6] Albanese A*, Lam AK*, Sykes EA, Rocheleau JV, Chan WC. Tumor-on-a-chip provides an optical window into nanoparticle transport, Nature Communications. 2013

Awards/Honors: Picower Fellowship Award, Institutional Award, 2017-2019 CIHR Postdoctoral Fellowship Award, National Award, 2015-2017 NSERC Postdoctoral Fellowship Award, National Award, 2014-2016 BRIAN ALBERTO AGUADO, PhD Chemical and , University of Colorado Boulder, 3415 Colorado Ave, UCB 596, Boulder, CO, 80303 . [email protected]

Research Overview: The long-term vision of the Aguado iBiomaterials Research Group will be to develop precision biomaterials that enable the evaluation of a patient’s unique biology to diagnose and treat a variety of health disorders. Recognizing the “one size fits all” approach to medicine is becoming increasingly outdated, I seek to exploit biomaterials as in vitro culture tools or implantable devices in vivo to understand how patient-specific variables (sex, age, and ancestry) may impact disease progression and treatments. With support from a NIH K99/R00 Pathway to Independence Award, my future lab will focus on sex as a biological variable and dissect molecular and cellular mechanisms governing sexual dimorphisms in cardiovascular disorders using precision biomaterials. Cardiovascular disorders, including aortic valve stenosis and cardiac fibrosis, are the leading cause of death in both men and women. However, the molecular and cellular mechanisms guiding cardiovascular disease progression in women remain poorly characterized because prior work rarely states the sex of cells used for in vitro studies or uses male animal models exclusively. To address this gap, we will pursue the following projects: 1. Determine intracellular X-linked and Y-linked molecular mechanisms that influence mechanotransduction and myofibroblast signaling pathways in cardiac using mechanically tunable hydrogels and next generation sequencing techniques. 2. Utilize sera from female and male patients with aortic valve stenosis to identify extracellular secreted factors and/or sex hormone compositions that contribute to persistent valvular myofibroblast activation during fibrosis. 3. Develop implantable polymer scaffolds that modulate recruited and resident macrophage phenotypes in female and male cardiac tissue to suppress fibrosis progression in small animal models. Taken together, my lab will fill an unmet need in understanding sexual dimorphisms in cardiovascular diseases using sex- specific engineered disease models. More broadly, our engineered tools will further enable and enhance our understanding of how patient-specific variables influence disease progression and treatments.

Education: B.S. Biomechanical Engineering, Stanford University, 2010 M.S. Biomedical Engineering, Northwestern University, 2013 Certificate in Management for Scientists and Engineers, Northwestern Kellogg School of Business, 2014 Ph.D. Biomedical Engineering, Northwestern University, 2015

Research/Work Experience: My postdoctoral research at the University of Colorado with Prof. Kristi Anseth identified serum proteins from aortic valve stenosis patients that mediate myofibroblast activation using in vitro disease models. In my future lab, I will use these models to probe intracellular (X/Y-linked) and extracellular (serum proteins, sex hormones) mechanisms guiding sex-specific myofibroblast activation. My doctoral research at Northwestern University with Prof. Lonnie Shea was partially focused on engineering biomaterial implants to manipulate macrophage phenotypes in primary tumors and suppress tumor cell invasion. In my future lab, I seek to use implants and small animal models to manipulate macrophage phenotypes during cardiac fibrosis, with the goal of slowing or halting fibrosis progression in a sex-specific manner.

Selected Publications: 1. Aguado BA et al. “Transcatheter aortic valve replacements alter circulating serum factors to mediate myofibroblast deactivation.” Science Translational Medicine, Accepted, in press, 2019. 2. Aguado BA et al. “Engineering precision biomaterials for personalized medicine.” Science Translational Medicine, 2018. 3. Aguado BA et al. “Biomaterial scaffolds as pre-metastatic niche mimics systemically alter the primary tumor and tumor microenvironment.” Advanced Healthcare Materials, 2018. *Cover article 4. Aguado BA et al. “Engineering the pre-metastatic niche.” Nature Biomedical Engineering, 2017. *Cover article

Awards/Honors: NIH K99/R00 Pathway to Independence Award (2019-2024) Burroughs Wellcome Fund Postdoctoral Enrichment Program Fellowship (2017-2020) NIH F32 NRSA Postdoctoral Fellowship (2017-2020) Howard Hughes Medical Institute Postdoctoral Fellowship (2016-2017) NSF Graduate Research Fellowship (2012-2015) HOSSEIN VAHID ALIZADEH, PhD Stanford University, 443 Via Ortega, Rm119, Stanford, California, 94305 [email protected]

Research Overview: My main research interests lie in Biomechatronics, particularly with respect to design and development of autonomous intelligent systems for medical applications. Biomechatronics is a combination of mechanical, electrical, and , sensor technology, artificial intelligence (AI), and Internet of Things (IoT). The majority of my research experiences have involved development of mechatronic, biomecial and robotic devices. My current postdoctoral research at Stanford University is focused on a revolutionary technology of concussion prevention. I am working on development of high-performance energy absorbers and integration of this technology into football helmets. At Stanford, I have also collaborated on a project on orienting oocytes using vibrations for In˗Vitro Fertilization (IVF) Procedures. In my previous postdoc experience at University of Toronto, I was involved in design and development of magnetically actuated surgical microrobots with fine tipped blades to be used in delicate brain surgeries or dissections. My doctoral dissertation examines theoretical and experimental approaches to the dynamical modeling and control of friction and wear in tribological systems via linear electromagnetic actuation. My PhD focuses on theoretical and experimental approaches to the real˗time closed˗loop control of frictional torque and lubrication by means of electromagnetic actuators (which provide a normal force to generate friction). In my M.Sc. dissertation, I utilized machine˗element design, dynamical modeling, real˗time closed˗loop control, image processing, path planning, and robot navigation to design and conduct the project “spherical mobile robot: theory and experiment.” In brief, I am most interested in taking a multi˗faceted approach to intelligent biomechatronic systems, spanning design, implementation, control, machine learning, performance evaluation, and integration into robotic and autonomous systems. My research interests are summarized below. Additional information and videos can be found here: http://www.hvahid.com/Research • BioMechatronics and : Biomechatronic systems, Autonomous systems, Surgical robotics, Navigation and control of robots, Vision-based motion control, Machine learning, Amphibious robots, Smart Structures • Bioengineering: Brain biomechanics, Biomechatronics, Brain injury mechanism, Concussion prevention systems • Dynamic Systems and Control: Automatic control, Real-time closed-loop control, Dynamical modeling, High frequency data acquisition, Vehicular and automotive technology, Automation, Robust and optimal control, feedback control theory

Education: • McGill University, 2017. PhD, Electrical and • University of Tehran, 2010 and 2007. MSc and BSc,

Research/Work Experience: • Stanford University, Bioengineering Department, 2018 - present Postdoctoral Researcher, Camarillo Lab, Advisor: Prof. Camarillo D. • University of Toronto, Mechanical and Department, 2017 Postdoctoral Researcher, Microrobotics Lab, Advisor: Prof. Diller E. • McGill University, Electrical and Computer Engineering Department, 2011 - 2017 Graduate Research Assistant, Control and Industrial Automation Laboratory, Centre for Intelligent Machines, Advisor: Prof. Boulet

Selected Publications: 1. Vahid Alizadeh H, Helwa MK, Boulet B. Modelling, Analysis and Constrained Control of the Wet Cone Clutch Systems: A Case Study of the Synchromesh System. Elsevier Journal. 49: 92-104 (2018) 2. Vahid Alizadeh H, Boulet B. Analytical calculation of the magnetic vector potential of an axisymmetric solenoid in the presence of iron parts. IEEE Transactions on Magnetics. 52(3): 1-4 (2016) 3. Vahid Alizadeh H, Helwa MK, Boulet B. Constrained control of the synchromesh operating state in an electric vehicle's clutchless automated manual transmission. IEEE Control Applications Conf. pp. 623-628 (2014) 4. Vahid Alizadeh H, Mahjoob MJ. Quadratic damping model for a spherical mobile robot moving on the free surface of the water. IEEE Symp. on Robotic and Sensors Environments. 125-130 (2011)

Awards/Honors: • Automotive Partnership Canada Award. 2014 - 2016 • McGill Engineering Doctoral Award. 2011 - 2014 • BMO Financial Group Major Fellowship. 2011 - 2014 HERDELINE ANN M. ARDOÑA, PhD Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138 [email protected] Research Overview: Taking inspiration from the macromolecular assembly structures in the biological millieu, I am interested in developing peptide-based supramolecular assemblies with engineered functional response to cellular interactions. My work as a graduate student involved the development of 1D peptidic nanostructures appended with organic electronic units, which exemplify tunable photophysical, mechanical and biochemical properties. This molecular design approach merges the biological relevance of self-assembling peptides and the optoelectronic function of organic semiconducting units—creating a bioelectronic scaffolding platform compatible with electrically-active cells such as and cardiomyocytes. The findings from this work enabled new ways to effectively generate localized electric fields within the internal electronic conduits of peptide-based nanomaterials. Semiconducting peptide monomers that can be assembled as 1D-nanostructures were successfully built according to design principles that allowed for directed photonic energy transport, sequential electron transport in a multicomponent system, and the transmission or equilibration of voltage or current when incorporated in a transistor device. In my postdoctoral work, I am continuing to investigate the multi-scale interactions of nanomaterials with biological systems. We are particularly exploring the nano-bio interface by understanding how engineered nanomaterials interact with biological barrier tissues (such as the endothelium and dermal layers) and how the biochemically entangled supramolecular networks of cytoskeletal proteins in barrier tissues reorganize in response to nanomaterial exposure. Moving forward, as an independent researcher, my goal is to build a research group that will pioneer next generation functional supramolecular assemblies that can be used as “adaptive probes” for monitoring biological processes such as cellular reorganization and signaling events. Developing bioinspired nanomaterials for investigating biological mechanisms opens the possibilities for unraveling new concepts governing cellular interactions at the biotic-abiotic interface.

Education: • Johns Hopkins University, August 2017. PhD, Chemistry • University of the Philippines Diliman, April 2011. BS, Chemistry, Summa Cum Laude

Research/Work Experience: • Harvard University, 2017-present Postdoctoral Fellow, Disease Biophysics Group, John A. Paulson SEAS and Wyss Institute; Advisor: Prof. Kit Parker • Johns Hopkins University, 2013-2017 Graduate Research Assistant, Department of Chemistry; Institute for Nanobiotechnology; and Translational Center-Johns Hopkins School of Medicine; Advisors: Profs. John D. Tovar and Hai-Quan Mao • University of the Philippines Diliman, 2009-2012 Instructor 5 (for General Chemistry and Organic Chemistry Laboratory), Institute of Chemistry (2011-2012) Undergraduate Researcher, Institute of Chemistry; Advisor: Prof. Susan D. Arco (2009-2011) • Avon Products Manufacturing Inc., Laguna, Philippines, April-May 2010. Intern, Quality Assurance Department.

Selected Publications: 1. S. Ahn, H.A.M. Ardoña, J. U. Lind, F. Eweje, S. L. Kim, G. M. Gonzalez, Q. Liu, J. F. Zimmerman, G. Pyrgiotakis, Z. Zhang, J. Beltran, B. Moudgil, P. Capinone, P. Demokritou and K.K. Parker. Mussel-inspired 3D fiber scaffolds for heart-on-a-chip toxicity studies of engineered nanomaterials. Anal. Bioanal. Chem. 410:6141 (2018) 2. H.A.M. Ardoña, E. R. Draper, F. Citossi, M. Wallace, L. Serpell, D.J. Adams, and J.D. Tovar. Kinetically controlled coassembly of multichromophoric peptide hydrogelators and the impacts on energy transport. J. Am. Chem. Soc. 139:8685 (2017) 3. H.A.M. Ardoña and J.D. Tovar. Peptide pi-electron conjugates: organic electronics for biology? Bioconjugate Chem. 26:2290 (2015) 4. K. Besar,* H.A.M. Ardoña,* J.D. Tovar and H.E. Katz. Demonstration of hole transport and voltage equilibration in self-assembled pi-conjugated peptide nanostructures using field-effect transistor architectures. ACS Nano 9:12401 (2015); *equal contribution 5. H.A.M. Ardoña, K. Besar, M. Togninalli, H.E. Katz and J.D. Tovar. Sequence-dependent mechanical, photophysical and electrical transport properties of pi-conjugated peptide hydrogelators.” J. Mater. Chem. C 3:6505 (2015) 6. H.A.M. Ardoña and J.D. Tovar. Energy transfer within pi-conjugated peptide heterostructures in aqueous environments. Chem. Sci. 6:1474 (2015)

Awards/Honors: • 12th Irving S. Sigal Postdoctoral Fellow, American Chemical Society (2018-2020) • International Student Research Fellowship, Howard Hughes Medical Institute (2015-2017) • Faculty for the Future Fellowship, Schlumberger Foundation (2014-2017) • Emmett and Elsie Buhle Fellowship Award, Johns Hopkins University (2014) • Leticia Shahani Award for Best Undergraduate Thesis, UP Diliman (2011) • Bank of the Philippine Islands-Department of Science and Technology: Science Award, Philippines (2010) • Baldomero M. Olivera, Jr. and Lourdes J. Cruz Award, UP Diliman (2010) PRABHANI U. ATUKORALE, PhD Dept. of BME, Case Western Reserve SOM, Case Comprehensive Cancer Center, Cleveland, OH 44106 (617) 922-0478 | [email protected]

Research Overview: I am a cancer immuno-bioengineer with a focus on the development of systemically administered immuno-nanoparticles that can home with high efficiency to the tumor microenvironment to deliver immune-potentiating agents that drive a powerful site-specific anti-tumor response. The generation of anti-tumor immunity at the tumor itself is pivotal to effective cancer vaccination, but is often absent in traditional lymph node-targeted vaccination strategies that solely aim to augment systemic immunity without addressing methods to recruit these immune cells to immunosuppressive tumor sites. Tumor-targeted immuno-nanoparticle vaccine strategies therefore represent a transformative paradigm shift. During my postdoctoral training with Prof. Efstathios Karathanasis (Case Comprehensive Cancer Center), I have developed an immuno-liposomal nanoparticle co-encapsulated with two synergistic immune agonists, targeting the STING and TLR4 pathways, that drains efficiently to the tumor perivasculature for uptake by sentinel antigen-presenting cells (APCs) in multiple animal models. These APCs, in turn, produce high levels of Type I interferon- to drive a local anti-tumor immune response that also serves to recruit systemic immune cells. Within this same context, I have also developed STING agonist-encapsulated mesoporous silica nanoparticles that not only drain to widely disseminated, clinically undetectable sites of dormant breast cancer metastasis to generate local anti-tumor immunity that results not only in tumor clearance but also in protective immunological memory against even isogenic cancers. This work has built on my foundational doctoral training with Prof. Darrell J. Irvine (MIT Koch Integrative Cancer Research Institute, HHMI), where my research focused on the development of ultra-small amphiphilic gold nanoparticles as cell membrane penetrating agents for delivery of vaccine antigens directly to the cell cytosol, bypassing endocytosis and priming a powerful CD8+ T cell response against aggressive melanoma.

Education: Massachusetts Institute of Technology, 2014. Ph.D., Bioengineering. Advisor: Prof. Darrell J. Irvine (HHMI) Johns Hopkins University, 2007. M.S.E., Biomedical Engineering. Advisor: Prof. Kevin J. Yarema Vanderbilt University, 2005. B.E., Biomedical Engineering, summa cum laude with Departmental Honors

Research/Work Experience: Case Western Reserve University SOM, 2014-Present. Postdoctoral Scholar. Supervisor: Prof. Efstathios Karathanasis Massachusetts Institute of Technology, 2007-2014. Graduate Research Assistant. Advisor: Prof. Darrell J. Irvine (HHMI) Johns Hopkins University, 2005-2007. Graduate Research Assistant. Advisor: Prof. Kevin J. Yarema Vanderbilt University, 2003-2005. Undergraduate Research Assistant. Honors Advisor: Prof. Adam W. Anderson

Selected Publications: 1. Atukorale PU, Raghunathan SP*, Raguveer V*, Moon TJ, Zheng C, Bielecki PA, Wiese ML, Goldberg AL, Covarrubias G, Hoimes CJ, Karathanasis E (2019). “Nanoparticle encapsulation of synergistic immune agonists enables systemic co-delivery to tumor sites and interferon -driven anti-tumor immunity.” Cancer Research. In press. 2. Atukorale PU, Guven SP*, Bekdemir A*, Carney RP*, Van Lehn RC, Yun DS, Jacob Silva PH, Demurtas D, Yang YS, Alexander-Katz A, Stellacci F, Irvine DJ (2018). “Structure-property relationships of amphiphilic nanoparticles that penetrate or fuse lipid membranes.” Bioconjugate Chemistry, 29(4):1131-40. 3. Atukorale PU, Yang YS, Bekdemir A, Carney RP, Silver PJ, Watson N, Stellacci F, Irvine DJ (2015). “Influence of the glycocalyx and plasma membrane composition on amphiphilic gold nanoparticle association with erythrocytes.” Nanoscale, 7(26):11420-32. 4. Yang YS, Atukorale PU*, Moynihan K*, Bekdemir A, Rakhra K, Tang L, Stellacci F, & Irvine DJ (2016). “Highthroughput quantitation of inorganic nanoparticle biodistribution at the single-cell level using mass cytometry.” Nature Communications, 8:14069. *Equal contribution 5. Van Lehn RC, Atukorale PU, Carney RP, Yang YS, Stellacci F, Irvine DJ, Alexander-Katz A (2013). “Effect of particle diameter and surface composition on the spontaneous fusion of monolayer-protected gold nanoparticles with lipid bilayers.” Nano Letters, 13(9):4060-7.

Selected Awards/Honors: Merck-Serono Innovation Cup Winning Team, Darmstadt, Germany, 2013 MIT Momenta Presidential Fellowship, 2007-2008 Johns Hopkins Medtronic Fellowship, 2005 Vanderbilt Robert H. McNeilly Memorial Engineering Honor Scholarship, 2002-2005 Vanderbilt Engineering Merit Scholarship (full tuition), 2001-2005 CHRISTINA M. BAILEY-HYTHOLT, PhD School of Engineering, Center for Biomedical Engineering, Brown University, 182 Hope St., Providence, RI, 02912 [email protected]

Research Overview: The focus of my research is advancing technologies for prenatal and women's health where I will leverage my expertise in biomaterials and diagnostics. Previously, I have led investigations on nanoparticle interaction studies and development of siRNA complexed micelles. As a NSF graduate research fellow, I then began my Ph.D. research towards bioengineering in women's health. The placenta is crucial during pregnancy, but remains one of the least understood organs. Placental trophoblast cells have great potential to provide information about pregnancy complications. During my Ph.D., I have developed a placental lipid bilayer model to investigate small molecule and environmental toxin interactions. There is little information on how pharmaceuticals and toxins interact with the maternal-fetal interface, and our system aims to improve this understanding. Additionally, we have developed a labelfree method to enrich trophoblast cells from clinical cervical samples using differential cell settling in solution. The capture of the entire fetal genome contained in these intact fetal trophoblast cells would be a significant improvement over current prenatal testing capabilities. As a future faculty member, I aim to (1) Enrich and isolate fetal exosomes from maternal samples for placental assessment and non-invasive diagnostics (2) Engineer the placental barrier and microenvironment for improved understanding of pregnancy-related complications and (3) Develop an 'artificial placenta' for improved nutrient transport to premature infants.

Education: Ph.D. in Biomedical Engineering, Brown University, Anticipated January 2020 B.S. in , Worcester Polytechnic Institute, 2015

Research/Work Experience: NSF Graduate Research Fellow, School of Engineering, Center for Biomedical Engineering, Brown University, 2015-Jan. 2020, Coadvisors: Dr. Anita Shukla and Dr. Anubhav Tripathi Undergraduate Researcher, Department of Chemical Engineering, Worcester Polytechnic Institute, 2012-2015, Advisors: Dr. Terri Camesano and Dr. Ramanathan Nagarajan (Natick Soldier Research, Development and Engineering Center)

Selected Publications: - Bailey-Hytholt, C.M., Puranik, T., Tripathi, A., Shukla, A., Impact of Environmental Toxicants on Lipid Structures, in preparation for Sept. 2019 submission. - Bailey-Hytholt, C.M., Sayeed, S., Kraus, M., Joseph, R., Shukla, A., Tripathi, A., A Rapid Method for Label-Free Enrichment of Rare Trophoblast Cells from Cervical Samples, Scientific Reports, 9, 12115, 2019. - Bailey, C.M., Tripathi, A., Shukla, A., Effects of Flow and Bulk Vesicle Concentration on Supported Lipid Bilayer Formation, Langmuir, 33(43), 11986-11997, 2017. - Bailey C.M., Nagarajan, R., Camesano, T.A., Designing polymer micelles of controlled size, stability and functionality for siRNA mdelivery, Control of Amphiphile Self-Assembling at the Molecular Level: Supra-Molecular Assemblies with Tuned Physicochemical Properties for Delivery Applications, pp.35-70, American Chemical Society, 2017 - Bailey, C.M., Kamaloo, E., Waterman, K.L, Wang K.F., Nagarajan, R., Camesano, T.A., Size dependence of gold nanoparticle interactions with a supported lipid bilayer: A QCM-D study, Biophysical chemistry, 203, 51-61, 2015. - Kamaloo, E., Bailey, C.M., Camesano, T.A., Effect of Concentration on the Interactions of Gold Nanoparticles with Model Cell Membranes: A QCM-D Study, Nanotechnology to Aid Chemical and Biological Defense (pp. 67-76), Springer, Dordrecht, 2015.

Awards/Honors: - National Science Foundation Graduate Research Fellowship, 2015-2020 - Biomedical Engineering Society Career Development Award, 2019 - Future Faculty Workshop, Princeton University, 2019 - Biomedical Engineering Society Travel Award, 2017 - 1st Place Poster Award, Central MA American Chemical Society, 2015 - 1st Place Poster Award, American Institute of Chemical Engineers, 2014 - Outstanding Women Student Awards (1 awarded annually), Worcester Polytechnic Institute, 2012 and 2014 AMAY J. BANDODKAR, PhD Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208 Email: [email protected]

Research Overview: My research interests lie in the development and characterization of soft, flexible, tissue-integrated devices for real-time monitoring of biochemicals with applications in wearable sweat sensing and neurochemical detection in live animals. These devices result from an unconventional amalgamation of disparate technologies, such as, lithography, laser patterning, electrochemical nano-structuring, enzymatic biofuel cells, colorimetric assays, advanced microfluidics, and wireless electronics. As a postdoctoral fellow at Northwestern University I developed the first example of wireless, self- powered sweat analysis and capturing systems that are several orders smaller and lighter than the most sophisticated sweat sensors reported to date. Separately, I developed a new class of integrated wireless, battery-free neural implants for simultaneous optical stimulation and neurochemical sensing in live mice. Studies with freely moving mice demonstrate the device’s capabilities to capture real-time dopamine transients under various conditions, such as, during optical stimulation, social interaction, and psychostimulant administration. As a PhD student at UC San Diego, I led my team in developing first examples of all-printed, highly stretchable, and self-healing devices for various wearable biosensing, energy harvesting, and energy storage applications. These devices can accommodate strains upto 500% and self-heal within seconds after damage. These experiences form the basis of my long-term research goals of developing unconventional soft, flexible biosensing systems for various wearable, implantable, and soft robotics applications that enable us to better understand the human physiology and develop tools for advanced human-machine interface.

Education: • University of California, San Diego, June 2016, PhD, Dept. of Nano-Engineering. • Indian Institute of Technology-Varanasi, India, May 2011, Integrated Masters, Dept. of Applied Chemistry.

Research/Work Experience: • Postdoctoral Fellow, Northwestern University, 2016 – present. • Summer Intern, Fraunhofer Institute for Biomedical Engineering, German, June 2010 – August 2010.

Selected Publications (Total Papers: 46; Total citations > 4050; h-index: 30): 1. “Soft, skin-interfaced microfluidic systems with passive galvanic stopwatches for precise chronometric sampling of sweat”, A. J. Bandodkar et al, Adv. Mater. 2019, 31, 1902109. 2. “Battery-free, skin-interfaced microfluidic/electronic system for simultaneous electrochemical, colorimetric & volumetric sweat analysis”, A. J. Bandodkar et al, Sci. Adv. 2019, 5, eaav3294. 3. “Soft, stretchable, high power density electronic skin-based biofuel cells for scavenging energy from human sweat” A. J. Bandodkar et al, Energy Environ. Sci., 2017,10, 1581. 4. “A wearable chemical–electrophysiological hybrid biosensing system for real-time health and fitness monitoring”, S. Imani, A. J. Bandodkar et al, Nat. Commun. 2016, 7, 11650.

Awards/Honors: 1. 2016 Materials Research Society Graduate Student Award. 2. 2016 Metrohm Young Chemist Award. 3. 2016 Siebel Scholars Award. 4. 2015 Interdisciplinary Research Award, UC San Diego. 5. NSF I-Corps Entrepreneurial Seed Fund, von Liebig Entrepreneurism Center (2015). 6. 2011 Undergraduate Research Publication Award. BENJAMIN I. BINDER-MARKEY, DPT, PhD Shirley Ryan Abilitylab & Northwestern University, Chicago, IL, 60618 . [email protected] Research Overview: The Edge Effect describes an ecologic phenomenon that occurs as 2 ecosystems meet. At this junction there exists more biodiversity and greater formation of new species than anywhere else on the planet. This “Edge Effect” is not isolated to just ecology but is present across all domains especially engineering and research. At the interaction points between specialties and areas, a diversity of perspectives and knowledge spur innovative, fresh, and impactful ideas that can be used to tackle the most difficult and pressing questions. Through my clinical training as a physical therapist and research experiences I have developed a unique set of skills and expertise across multiple domains to take advantage of the Edge Effect. I my research lives at the “edge” between physical therapy and engineering, the nervous system and muscles, experimental and modeling, and the micro and macro scales. My research aims to answer the complex question of how adapts after a neural injury and how these adaptations interact with the impaired nervous system preventing optimal recovery. These interactions are complex. Therefore, I use complementary experimental, imaging, and computational musculoskeletal modeling methods to elucidate the consequences of the muscle and neural impairments that would otherwise be difficult to distinguish using one method alone. To understand how skeletal muscle adapts, I explore the adaptations across size scales, from the microscopic scale of cellular muscle fibers to the macroscopic scale of joint and whole-body mechanics. Looking across scales gives novel insights into how adaptations across the scales interact with each other to affect function. Finally, with an understanding of these adaptations and interactions, therapeutic interventions and devices can be developed preventing or reversing these impairments, allowing individuals to achieve their optimal recovery.

Education: PhD, Biomedical Engineering, 2018, Northwestern University; Advisors: Drs. Wendy M Murray and Julius PA Dewald DPT, Doctorate of Physical Therapy, 2015, Northwestern University B.E., Mechanical Engineering, 2008, University of Delaware; Advisor: Dr. Thomas Buchanan

Research/Work Experience: 2018- Present: Post-Doctoral Fellow. Shirley Ryan AbilityLab (Formerly the Rehabilitation Institute of Chicago) & Northwestern University, Department of Physical Medicine and Rehabilitation; Mentors: Dr. Richard Lieber, PhD & Dr. Elliot Roth, MD 2008 – 2009: BioImaging Technician. University of Delaware, Department of Mechanical Engineering

Selected Publications: Binder-Markey BI, Broda N, Lieber RL: Intramuscular anatomy drives muscle collagen content variation within and between muscles – In Preparation Binder-Markey BI, Murray WM, Dewald JPA: The effect of altered neural inputs and botulinum neurotoxin chemical denervation on the passive biomechanical properties of hand and wrist muscles in chronic hemiparetic stroke – In Preparation Binder-Markey BI, Dewald JPA, Murray WM: The sensitivity of the development of the claw finger deformity to biomechanical changes of the finger after intrinsic muscle paralysis: A computational biomechanical model study – Journal of Hand Surgery, 2019 (In Press) Binder-Markey BI, Murray WM: Incorporating the length-dependent passive-force generating muscle properties of the extrinsic finger muscles into a wrist and finger biomechanical musculoskeletal model, Journal of Biomechanics, 2017 Aug, 61:250-7 Binder-Macleod BI, Buchanan TS: Tibialis anterior volumes and areas in ACL-injured limbs compared with unimpaired, Med Sci Sports Exerc. 2006 Sep;38(9):1553-7.

Awards/Honors: ·Post-Doctoral Fellowship in Stroke Research, 2018-2020, Shirley Ryan AbilityLab & Brinson Foundation ·Pre-Doctoral Fellowship, 2016 – 2018, American Heart Association ·Doctoral Student Presentation Competition Finalist, 2018, American Society of Biomechanics Annual Meeting ·ASME-BED PhD Level Student Paper Competition Runner-up, 2018, World Congress of Biomechanics ·R. W. Jones Research Award in Neural Engineering and Rehabilitation, 2017, Northwestern Department of Biomedical Engineering ·Promotion of Doctoral Studies (PODS) II Scholarship, 2016 – 2017, Foundation for Physical Therapy ·Mary Lou Barnes Award, 2016, American Physical Therapy Association Section on Neurology ·Dean’s Feinberg DPT/PhD Scholar, 2015, Northwestern University Department of Physical Therapy and Human Movement Sciences SUMAN BOSE, PhD Koch Institute for Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139 [email protected]

Research Overview: My broad research interest is in applying engineering approaches to study, model, and manipulate complex biological systems for curing disease. To enable this, I will develop high-throughput microfluidic platforms that can perform multidimensional quantitative measurements with single-cell resolution. I will use this to gain fundamental insights into how cells functions and interacts within their complex environment, and how dysregulation of these processes can lead to diseases such as cancer. Another focus area will be engineering implantable devices that can manipulate the local environment within the body for cell- therapy and tissue engineering application. We are interested in applying this technology to develop new therapies for diabetes and chronic autoimmune diseases. I am currently an NIH Pathway to Independence (K99) fellow in the laboratories of Prof. Robert Langer and Daniel Anderson at MIT. Funded by a JDRF Postdoctoral Fellowship, I have developed a biocompatible cell-encapsulation macrodevice that can effectively transplant immunogenic therapeutic cells in animals. I discovered that membrane porosity could modulate the infiltration of immune cells in vivo and also developed an effective surface coating that can mitigate device fibrosis. My current research, in collaboration with Prof. Phillip Sharp, has been to develop a microfluidic platform to enable high throughput sequencing of microRNA and other regulatory RNAs from single cells. I am using the platform to study miRNA regulation during differentiation of embryonic stem cells, and study cell heterogeneity from cancer patients.

Education: Ph.D., Massachusetts Institute of Technology, 2014 M.S, Massachusetts Institute of Technology, 2009 B.Tech, Indian Institute of Technology Kharagpur, India, 2007

Research/Work Experience: 2018 - | Research Scientist/K99 Fellow, MIT. Mentors: Daniel Anderson, Robert Langer, and Phillip Sharp. Research focus: Single-cell genomics, miRNA profiling of cancer cells. 2014 - 2018 | JDRF Postdoctoral Fellow, MIT. Adviser: Daniel Anderson and Robert Langer Research focus: Implantable immune-protective macrodevice for cell-based therapy. 2007-2014 | Graduate Research Assistant, Mechanical Engineering Department, MIT. Adviser: Prof. Rohit Karnik. Research focus: Microfluidic cell-sorting platform for diagnostic applications.

Selected Publications: Bose S, Volpatti L, Thiono D, Yesilyurt V, McGladian C, Wang A, Weir G, Langer R, Anderson DG, A biocompatible macrodevice enables long-term survival of therapeutic xenogeneic cells in vivo. Nature Biomedical Engineering (in final revision) Bose S, Singh S, Hollatz M, Shen C, Lee CH, Dorfman DH, Karp JM, Karnik R, Affinity flow fractionation of cells via transient interactions with asymmetric molecular patterns, Scientific Reports, 3, 2013. Zhao W*, Cui C*, Bose S*, Guo D, Shen C, Wong W, Halvorsen K, Farokhzad O, Teo G, Phillips J, Dorfman D, Karnik R, Karp, JM, A bioinspired multivalent DNA network for capture and release of cells, PNAS, 109(48), 2012. Lee CH*, Bose S*, VanVillet K, Karp JM, Karnik R, Examining the lateral displacement of HL60 cells rolling on asymmetric PSelectin patterns, Langmuir, 27(1), 2011. Bose S, et.al., A semianalytical model to study the effect of cortical tension on cell rolling, Biophysical Journal, 99(12), 2010.

Awards/Honors: · NIBIB/NIH Pathway to Independence Award (K99/R00), 2018-23 · Keystone Future of Science Award, 2017 · Top 40 under 40 Healthcare Innovators, MedTech Boston, 2016 · KI Mission Possible Grand Prize ($300K), 2016 · JDRF Postdoctoral Fellowship 2015-18 · Pappalardo Fellowship, MIT, 2007 · Institute Silver Medal, IIT Kharagpur, 2007 ANNIE C. BOWLES, PhD 1Biomedical Engineering, University of Miami, 1951 NW 7th Ave 475, Miami, FL, 33136, 2Orthopaedics, University of Miami, 1450 NW 10th, 3012, Miami, FL, 33136 [email protected]

Research Overview: Decades of evidence demonstrate the unique capabilities and robust therapeutic effects of mesenchymal stem/stromal cells (MSCs), however researchers are now able to recognize limitations to the standard protocols, including isolation and culture, that hinder clinical translatability. In short, isolation techniques generate “crude” batches of MSCs composed of heterogeneous subpopulations, and two-dimensional cultures of adherent MSCs are diminishing the overall biological properties of the cells. Together, these methodsintroduce inconsistencies in outcomes that confound evidence-based achievements for regulatory compliance, and ultimately limit the advancement of promising candidate biologics for numerous clinical indications. My research proposes improved techniques to overcome these limitations by several approaches and has acquired robust evidence that demonstrates the enhanced quality of MSCderived\ effects. Moreover, selection of high quality MSCs by conducting in-depth potency assays help to identify potential universal donors and associate donor demongraphics with therapeutic performance.

Education: Postdoctorate in Biomedical Engineering and Orthopedics, present, University of Miami, Miami, FL Doctor of Philosophy, 2017, Tulane University, New Orleans, LA Master of Science, 2012, Tulane University, New Orleans, LA Bachelor of Science, 2008, Louisiana State University, Baton Rouge, LA

Research/Work Experience: Cell and Molecular Biologist with over 8 years of experience evaluating Mesenchymal Stem/ (MSC) based therapeutics applied to various fields of science and industry with emphasis in: • MSC physiology and immunomodulation • Cell manufacturing • Biologics and cell-free products (exosomes) • Small business consulting • Immunology, autoimmunity, neurodegeneration • Molecular Orthopedics and Orthobiologics • MSCs for co-transplantation with for • Organ-on-chip devices and applications • Wound healing and skin rejuvenation, Breast cancer progression and metastasis

Selected Publications: Bowles AC, Wise RM, Gerstein BY, Thomas RC, Ogelman R, Manayan RC, Bunnell BA. (2018) Adipose Stromal Vascular Fraction Attenuates TH1 Cell-Mediated Pathology in a Model of Multiple Sclerosis. Journal of Neuroinflammation, 15(1):77. PMID: 29534751. Bowles AC, Wise RM, Gerstein BY, Thomas RC, Ogelman R, Febbo I, Bunnell B.A. (2017). Immunomodulatory Effects of Adipose Stromal Vascular Fraction Cells Promote Alternative Activation Macrophages to Repair Tissue Damage. Stem Cells, 35:2198-2207. PMID: 28801931 Bowles AC, Strong AL, Wise RM, Thomas RC, Dutreil MF, Hunter RS, Gimble JM, Bunnell BA. (2016). Adipose Stromal Vascular Fraction-Mediated Improvements at Late Stage Disease in a Murine Model of Multiple Sclerosis. Stem Cells, 35(2):532-544. PMID: 27733015

Awards/Honors: 2019 Grant Awardee, National Institute of Health Loan Repayment Program 2018 Postdoctoral Shooting Star Award, Cellular and Molecular Bioengineering Conference, Biomedical Engineering Society, San Diego, CA 2017 Travel Award for Conference, Human Islet Research Network, Cambridge, MA 2017 Best Poster Award, Executive Advisory Board meeting for JTMF Biomedical Nanotechnology Institute at University of Miami 2016 James de la Houssaye Mentor Award, Greater New Orleans Science and Engineering Fair LAURA G. BRACAGLIA, PhD Biomedical Engineering, Yale University, 55 Prospect Street, New Haven, Connecticut, 06511 [email protected]

Research Overview: My research goal is to design tissue-engineered scaffolds with built-in tools for long-term immune modulation. Applying these two fields to site-specific disease management could add greatly to the applicability of regenerative medicine. My research goal is grounded in the fields of tissue engineering and biomaterial scaffolds from my graduate training at the University of Maryland. In my thesis work, I developed several iterations of biohybrid materials, combining man-made polymers with natural biomaterials, to be used for cardiovascular surgical reconstruction. We found that the inclusion of cardiovascular molecules in scaffolds supported cell regrowth, while the inclusion of biodegradable polymers allowed for the control of shape (through 3D-printing), strength, degradation rate, and could serve as a drug delivery source to reduce inflammation or injury. In my postdoctoral research at Yale, I have studied more specifically direct manipulations of the immune system interaction with dysfunctional endothelial cells. My research is aimed at the development of nanoparticle-based therapies which can be targeted to the endothelium for the purpose of preventing inflammatory related damage. Through the sustained release of nucleic-acid or small molecule delivery from novel polymeric systems to regionally targeted endothelial cells in intact human vessels, the immune cell activation and propagation of several inflammatory pathways is reduced. We are actively applying this strategy to prevent immune- related injury in solid . Leveraging these training and investigative experiences, my faculty research plan will focus on developing implantable scaffolds to replace disease- or injury-damaged vasculature. An implantable device that not only supports the growth of new tissue but also directs healing mechanisms or the suppression of injury- causing immune pathways would broaden the impact of regenerative medicine beyond the regrowth of tissue to site- specific immune inhibition or to resolve continued injury.

Education: Ph.D. Bioengineering, University of Maryland May 2017 B.S. Biomedical Engineering, Georgia Institute of Technology May 2012

Research/Work Experience: Postdoctoral Fellow (since July 2017) Department of Biomedical Engineering, Yale University, Advisor: W. Mark Saltzman, PhD Graduate Research Assistant (2012-2017) Fischell Department of Bioengineering, UMD, Advisor: John P. Fisher, PhD Visiting Graduate Research Assistant (2015) SFI Centre for Research in Medical Devices (CÚRAM) NUIG Research Intern (2012) FDA Center for Device and Radiological Health, Division of Solid and Fluid Mechanics Undergraduate Research Assistant (2009-2012) Cardiovascular Fluid Mechanics Laboratory, Georgia Institute of Technology NREIP Research Intern (2010-2011) US Naval Research Laboratory Center for Biomolecular Science and Engineering

Selected Publications: 1. Cui, J., Piotrowski-Daspit, A. S., Zhang, J., Shao, M., Bracaglia, L. G., Utsumi, T., Seo, Y. E., DiRito, J., Song, E., Wu, C., Inada, A., Tietjen, G. T., Pober, J. S., Iwakiri, Y., Saltzman, W. M., Poly(Amine-co-Ester) Nanoparticles For Effective Nogo-B Knockdown in the Liver. J Control Release 2019, 304, 259-267. 2. Tietjen, G.T., Bracaglia, L.G., Saltzman, W.M., Pober, J.S. Focus on Fundamentals: Achieving Effective Nanoparticle Targeting. Trends in molecular medicine, 2018 3. Bracaglia, L. G.; Winston, S.; Powell, D., Fisher, J. P., Synthetic Polymer Coatings Diminish Chronic Inflammation Risk in Large ECM-Based Materials. J Biomed Mater Res A 2019 4. Bracaglia, L. G.; Messina, M.; Winston, S.; Kuo, C. Y.; Lerman, M.; Fisher, J. P., 3D Printed Pericardium Hydrogels to Promote Wound Healing in Vascular Applications. Biomacromolecules 2017, 18 (11), 3802-3811. 5. Bracaglia, L. G.; Messina, M.; Vantucci, C.; Baker, H. B.; Pandit, A.; Fisher, J. P., Controlled Delivery of Tissue Inductive Factors in a Cardiovascular Hybrid Biomaterial Scaffold. ACS Biomaterials Science & Engineering 2017, 3 (7), 1350-1358.

Awards/Honors: 2016 NIH Ruth L. Kirschstein National Research Service Award (F31) Predoctoral Fellowship 2015 University of Maryland International Graduate Student Research Award 2014 American Heart Association Individual Predoctoral Fellowship 2014 The Mary Ann Liebert, Inc. Outstanding Student Award, TERMIS 2016 TERMIS, Chair of Student and Young Investigator Section, Americas Chapter (elected role) 2015 University of Maryland Graduate School All-S.T.A.R. Fellowship DOUGLAS K. BRUBAKER, PhD Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139 [email protected]

Research Overview: Pre-Clinical to Clinical Translational Systems Biology of Inflammatory Pathologies and Cancer A fundamental challenge in translational biomedicine is the prediction of when molecular or phenotypic responses in one biological context hold true in another. This challenge underpins efforts at forward translation from model systems to humans, reverse translation from clinical cohorts to experimental studies, and translation between clinical cohorts. Despite considerable efforts to engineer mouse and cell culture systems more similar to human physiology, the failure rate for therapeutic and mechanistic hypothesis translation remains high. The principle behind my research is that translating systems models of signaling, rather than individual observations, will catalyze more impactful translation of preclinical biology to improve the understanding and treatment of complex diseases. The goal of the Brubaker Lab will be to understand how dysregulation of tissue, immune, and microbial signaling manifests in disease and to apply this understanding to develop therapeutic strategies to correct disease signaling in complex inflammatory diseases and cancers. To achieve this goal, my lab will develop new computational methods to translate molecular insights between model systems and patients, enabling us to integrate information from multiple molecular and clinical data modalities to uncover links between therapeutic response, cell-type specific, and host-microbiome signaling. We will explore these hypotheses through biological experiments based on our own expertise or in collaboration with other experimental biologists. We will also seek collaborations with basic scientists and clinicians to apply our approaches to translational challenges in other disease contexts. My laboratory’s research areas will be 1) Translational Pharmacogenomics of Cancer and Inflammatory Disease Subtypes, 2) -Specific Signaling Translation, and 3) Host-Microbiome Signaling Translation. These complementary, but distinct, research areas are a synthesis of my graduate and postdoctoral experience developing new computational tools to understand and correct signaling dysregulation. Initially, my lab will pursue questions in inflammatory bowel diseases and colorectal cancer, ideal model diseases driven by a combination of genetic, environmental, immune, microbial, and host tissue signaling factors. We will develop subtype-tailored therapeutic hypotheses based on multi-omic models of patient and experimental datasets. Integrating subtypespecific signaling models with single-cell RNA-sequencing data will allow us to identify cell type-specific therapeutic resistance mechanisms. Combining subtype and cell type-specific signaling models with microbial and metabolomics data will reveal interactions between microbial species and host cell types driving disease progression and therapeutic resistance. We will test these hypotheses in transwell-based in vitro culture systems of intestinal and immune cells to model the gut-immune axis and use stimulation with microbial products to model the interaction of intestinal and immune cells with the microbiome. Altogether, our work will provide a systems view of dysregulated host, immune, and microbial signaling to catalyze discovery of new therapeutic strategies.

Education: Ph.D. Systems Biology and Bioinformatics 2016, Case Western Reserve University B.S. Mathematics 2013, Case Western Reserve University

Research/Work Experience: Post-doctoral Research Associate (2016-Present). Massachusetts Institute of Technology Biological Engineering, Boehringer Ingelheim Pharmaceuticals Research Beyond Borders, and Harvard Medical School Beth Israel Deaconess Medical Center. Graduate Student Researcher (2013-2016). Case Western Reserve University Center for Proteomics and Bioinformatics.

Selected Publications: 1. Brubaker D.K.* and Paulo J.A.,* et al., “Proteogenomic network analysis of context-specific KRAS signaling in mouse-to-human cross-species translation.” Cell Systems. (2019). *Co-First Author. 2. Brubaker D.K., Proctor E.A., Haigis K.M., Lauffenburger D.A., “Computational translation of genomic responses from experimental model systems to humans.” PLoS Computational Biology. (2019). 3. Lyons J.* and Brubaker D.K.,* et al., “Integrated in vivo multiomics analysis identifies p21-activated kinase signaling as a driver of colitis.” Science Signaling. (2018). *Co-First Author.

Awards/Honors: Speaker Travel Award Keystone Meeting on The Resolution of Inflammation in Health and Disease. 2018. Speaker Travel Award AAAS Science Signaling, Interdisciplinary Signaling Workshop. 2017. Speaker Travel Award Society of Industrial and Applied Mathematics, Applications of Dynamical Systems. 2015. ALEXANDER BUFFONE, Jr., PhD 1Bioengineering, University of Pennsylvania, 540 Skirkanich Hall, 220 S 33rd Street, Philadelphia, PA, 19104, 2Surgery, University of California-San Francisco, 513 Parnassus Avenue-560 HSE, San Francisco, CA, 94143 [email protected]

Research Overview: The Buffone Lab: Perturbing the immune and cancer cell glycocalyx to tune migration The glycocalyx, or the large network of sugar coated glycoproteins, glycolipids, and glycosaminoglycans which cover the surface of all cells, plays a role in cellular signaling, proliferation, and most critically migration. Controlling the content and the structure of these glycans is of critical therapeutic interest as they control the ability of both immune cells migrating to the sites of inflammation and cancer cells metastasizing to secondary organs to reach their intended targets. Thus the ability to discover any modify critical components of the glycocalyx allow for the engineering of: immune cells to better reach the sites of and resolve inflammation and cancer cells to prevent their spread and through the blood stream and the formation of secondary tumors. The Buffone Lab will take a global view of immune cell and cancer cell migration and will focus on controlling the cells migratory ability through modifications to their glycocalyx by: 1) Identifying the critical glycosyltranferases and ligands which promote greater selectin mediated adhesion; 2) Perturbing and defining the roles of N-linked, O-linked and glycolipid synthesis in mesenchymal like tumor cells 3) Assaying for tumor cells which may migrate against the direction of shear flow along the cellular adhesion molecules (CAMs) presented on the endothelial surface during metastasis.

Education: •State University of New York at Buffalo, May 2012. PhD, Chemical Engineering •State University of New York at Buffalo, June 2006. BS, Chemical Engineering

Research/Work Experience: •Visiting Scholar (February 2019-Present), University of California, San Francisco (San Francisco, CA), Department of Surgery, Advisor: Prof. Valerie Weaver. Postdoctoral Fellow (September 2015-January 2019) and Research Associate (February 2019-Present), University of Pennsylvania (Philadelphia, PA), Department of Bioengineering, Advisor: Prof. Daniel Hammer. •Postdoctoral Fellow (September 2012-August 2015), Roswell Park Cancer Institute (Buffalo, NY), Department of Molecular and Cellular Biology, Advisor: Prof. Joseph Lau. •Graduate Research Assistant/PhD Student (September 2006-July 2012), State University of New York at Buffalo (Buffalo, NY), Department of Chemical and Biological Engineering, Advisor: Prof. Sriram Neelamegham. •Undergraduate Researcher (June 2005 – May 2006), SUNY Buffalo (Buffalo, NY), Department of Chemical and Biological Engineering, Advisor: Prof. Mattheos Koffas.

Selected Publications: 1) Buffone A Jr., Anderson NR, Hammer DA. “Human neutrophils will crawl upstream on ICAM-1 if Mac-1 is blocked.” Biophys J. 2019, Accepted 2) Buffone A Jr., Anderson NR, Hammer DA. “Migration against the direction of flow is LFA-1 dependent in human hematopoietic stem cells.” J Cell Sci. 2018 131: jcs205575. Epub 17 Nov 27 3) Buffone A Jr., Nasirikenari M, Manhardt, CT, Lugade A, Bogner PN, Sackstein, R., Thanavala Y, Neelamegham S, Lau JT. “Leukocyte-borne α(1,3) fucose is a negative regulator of β2 integrin-dependent recruitment in lung inflammation.” J Leukoc Biol. 2017 Feb;101(2):459-470. Epub 2016 Aug 26. 4) Mondal N*, Buffone A Jr*, Stolfa G, Antonopoulos A, Lau JT, Haslam SM, Dell A, Neelamegham S. “ST3Gal-4 is the primary sialyltransferase regulating, E-, P-, and L-selectin ligands on human myeloid leukocytes.” Blood. 2015 Jan 22; 125(4):687-96. Epub 2014 Dec 10. *Co-First Authors 5) Buffone A Jr., Mondal N, Gupta R, McHugh KP, Lau JT, and Neelamegham S. “Silencing α1,3-fucosyltransferases in human leukocytes reveals a role for FUT9 enzyme during E-selectin mediated cell adhesion.” J Biol Chem. 2013 Jan 18; 288(3):1620-33. Epub 2012 Nov 28.

Awards/Honors: •Co-investigator on R21 which I wrote from NIGMS titled: “Controlling the Upstream Migration of Neutrophils through the modulation of Mac-1”. Project dates: (7/1/19-6/30/21) •Research Highlighted in a BMES Blog Post, 2018 •Highlighted in a First Person Interview in Journal of Cell Science, 2018 •Society for Glycobiology Travel Award Recipient, 2014 •University at Buffalo CBE Department Open House Speaker, 2011 MARK A. CALHOUN II, PhD Biomedical Engineering, , 308 Research dr, C-138, Durham, North Carolina, 27708 [email protected]

Research Overview: My research program will develop new tools that enable us to harness differential signaling induced by stimuli in the neural microenvironment for improved nerve injury and cancer treatment. I will leverage my experience in mechanobiology, microfluidics, molecular biology, and device design to develop in vivo devices to augment the diseased microenvironment and to build biomimetic in vitro models of microenvironment stimuli. In my current work with Ravi Bellamkonda at Duke University, I have been particularly interested in the intersection of electric fields, peripheral nerve injury, and senescence. Using microfluidic and molecular biology techniques, we are studying how Schwann cell repair phenotypes (i.e. migration, proliferation, neurotrophin expression) are altered in response to a novel electric field waveform. In parallel, we have developed an implanted in vivo device to deploy this electric field in vivo. Senescence is a key facet of the peripheral nerve pathology and is implicated in poor nerve repair. By seeking to understand how it is involved in and modulated by electric fields, we advance both the engineering and the biology. Ultimately, this line of research will result in devices that augment the neural microenvironment to enhance repair in different kinds of trauma to the nervous system. In my graduate work with Jessica Winter and Jose Otero at The Ohio State University, I was focused on the intersection of mechanobiology, biomaterials, and glioblastoma. Using electrospun fiber mats and custom in vitro biomimetic models, we explored the effects of physical stimuli in the glioblastoma microenvironment on tumor progression phenotypes (e.g. invasion, survival, etc.). These physical stimuli were induced by tumor progression and included interstitial fluid pressure, substrate stiffness, and mechanical compression. Specifically, we showed how elevated mechanical compression induced increased motility and was predicted to decrease proliferation. Those changes correlated with miR-548 family regulation, which we highlighted as a candidate molecule for pharmaceutical intervention. The long-term goal of this work is to identify and leverage differential signaling induced by the microenvironment to improve the dismal prognosis of glioblastoma patients. Collectively, my research program will involve device development to deploy desired stimuli in vivo countered with building in vitro models to study the effects of stimuli from the microenvironment. This will highlight candidate drug targets, advance the biology of cell response to extracellular cues, and yield novel devices and therapies that improve patient outcomes in nerve injury and repair. ÷÷÷÷

Education: 2017 Ph.D., Biomedical Engineering, The Ohio State University 2012 B.S., Biomedical Engineering, Rose-Hulman Institute of Technology

Research/Work Experience: 2017-Present: Postdoctoral Associate, Duke University, Bellamkonda Lab 2012-2017: Graduate Fellow, The Ohio State University, Winter Lab

Selected Publications: Calhoun, MA*; Cui, Y*; Elliott, E; Mo, X; Otero, J; Winter, J. MicroRNA-mRNA Interactions at Low Levels of Compressive Solid Stress Implicate mir-548 in Increased Glioblastoma Cell Motility. Submitted to Scientific Reports. * Equal Contributors. Calhoun, M.; Bentil, S. A.; Elliott, E.; Otero, J. J.; Winter, J. O.; Dupaix, R. B. Beyond Linear Elastic Modulus: Viscoelastic Models for Brain and Brain Mimetic Hydrogels. ACS Biomater. Sci. Eng. 2019, DOI: 10.1021/acsbiomaterials.8b01390 Calhoun MA, Chowdhury SS, Nelson MT, Lannutti JJ, Dupaix RB, Winter JO. Effect of Electrospun Fiber Mat Thickness and Support Method on Cell Morphology. Nanomaterials (Basel). 2019;9(4):644. Published 2019 Apr 20. doi:10.3390/nano9040644 Nabar GM, Mahajan KD, Calhoun MA, et al. Micelle-templated, poly(lactic-co-glycolic acid) nanoparticles for hydrophobic drug delivery. Int J . 2018;13:351–366. Published 2018 Jan 10. doi:10.2147/IJN.S142079

Awards/Honors: Funding: 2015-2017 Pelotonia Graduate Research Fellowship 2014-2015 HHMI Med into Grad Fellowship 2013 Nanoscale Science and Engineering Center Fellowhip Academic (selected): 2019 Rising Stars in Biomedical 2017 2nd Place Oral Presentation, Hayes Graduate Research Forum, OSU 2016 Chair's Award for Outstanding and Exceptional Contribution to BME LESLIE CHAN, PhD Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, Rm 76- 453, Cambridge, Massachusetts, 02139 . [email protected]

Research Overview: The focus of my research is the development of nanoscale materials that interact with tissue microenvironments for diagnostic and therapeutic applications. In my current work, I have engineered a diagnostic platform for respiratory disease, which consists of multiplexable, volatile-releasing nanoparticles. These nanoparticles are delivered into the lungs to generate a breath readout for aberrant protease activity. By pairing this platform with cost-effective volatile detectors and machine learning, the goal is to generate disease-specific volatile signatures for disease detection at the point of care. I am currently applying this platform in bacterial and viral pneumonia, alpha-1 antitrypsin deficiency, and lung cancer. By leveraging volatile trafficking in the body, my future goals include adapting this platform for extrapulmonary diseases.

Education: 2010-2015 University of Washington, Seattle, WA / Doctor of Philosophy in Bioengineering 2006-2009 Georgia Institute of Technology, Atlanta, GA / Bachelor of Science in Biomedical Engineering

Research/Work Experience: 2015-Present Postdoctoral Associate, Massachusetts Institute of Technology, Cambridge, MA Postdoctoral Advisor: Professor Sangeeta Bhatia Koch Institute for Integrative Cancer Research Engineering a diagnostic platform for detecting and monitoring respiratory diseases through a breath readout for protease activity in the lungs. 2010-2015 Graduate Research Assistant, University of Washington, Seattle, WA Graduate Advisor: Professor Suzie Pun Department of Bioengineering Engineered synthetic polymer materials for in situ crosslinking of fibrin matrices to accelerate blood clotting for hemostatic applications. 2007-2009 Undergraduate Research Assistant, Georgia Institute of Technology, Atlanta, Georgia Advisor: Professor Ravi Bellamkonda Department of Biomedical Engineering Developed multifunctional liposomal carriers containing CT/MRI contrast agents and doxorubicin for interrogation of tumor vascular permeability and monitoring of drug delivery.

Selected Publications: 1. L. W. Chan, N. J. White, S. H. Pun, A Fibrin Cross-linking Polymer Enhances Clot Formation Similar to Factor Concentrates and Tranexamic Acid in an in vitro Model of Coagulopathy, ACS Biomaterials Science and Engineering 2, 403-408 (2016). 2. L. W. Chan, X. Wang, H. Wei, L. D. Pozzo, N. J. White, S. H. Pun, A Synthetic Fibrin-Crosslinking Polymer for Modulating Clot Properties and Inducing Hemostasis, Sci. Transl. Med. 7, 277ra29 (2015). 3. L. W. Chan, N. J. White, S. H. Pun, Synthetic Strategies for Engineering Intravenous Hemostats, Bioconjug. Chem. 26, 1224–1236 (2015). 4. L. W. Chan, C. H. Kim, X. Wang, S. H. Pun, N. J. White, T. H. Kim, PolySTAT-modified Chitosan Gauzes for Improved Hemostasis in External Hemorrhage, Acta Biomaterialia 31, 178-85 (2015). 5. L. W. Chan, Y. Wang, L. Y. Lin, M. P. Upton, J. H. Hwang, S. H. Pun, Synthesis and Characterization of Anti-EGFR Fluorescent Nanoparticles for Optical Molecular Imaging, Bioconjug. Chem. 24, 167–175 (2012).

Awards/Honors: 2019-2023 NIH K99/R00 Pathway to Independence Award 2019 Caltech Young Investigator Lecturer in Engineering and Applied Science 2018-2019 Marble Center Convergence Scholars Program (CSP) 2018 BMES Career Development Award 2015 UW College of Engineering Award for Outstanding Service and Achievement 2013-2015 Bioengineering Cardiovascular Training Grant Fellowship (T32) 2012-2013 Nanotechnology and Physical Science in Cancer Research Fellowship (T32) CRYSTAL K. CHE, PhD Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139 [email protected]

Research Overview: Organic chemistry, applied to the preparation of small molecules and polymers, is a central science for the development of functional materials. My research program will specifically address the molecular design of biomaterials and elucidation of structure-property relationships to be utilized for unexplored applications and production methods. During my graduate training, I established new reaction methodologies and design principles for catalyst development. In my postdoctoral research, I am applying these fundamental concepts from organic chemistry to develop rational design strategies of polymeric materials with tunable properties for drug delivery and tissue engineering. I will continue a molecular approach to establish structure-property relationships for organic materials design in the next stage of my career, and am excited to mentor students and postdocs in a program dedicated to research at the intersection of organic chemistry, materials science, and biomedical engineering with the goal of addressing longstanding challenges in medicine.

Education: Ph.D. in Chemistry, 2012–2017, California Institute of Technology B.S. in Chemistry, 2008–2012, University of California, Berkeley

Research/Work Experience: NIH Postdoctoral Fellow/Postdoctoral Associate, 2017–present, Massachusetts Institute of Technology, Koch Institute for Integrative Cancer Research, Advisors: Professor Robert S. Langer and Professor Daniel G. Anderson Graduate Research Assistant, 2012–2017, California Institute of Technology, Division of Chemistry and Chemical Engineering, Advisors: Professor Robert H. Grubbs and Professor Gregory C. Fu Undergraduate Research Assistant, 2009–2012, University of California, Berkeley, College of Chemistry, Advisors: Professor Jean M. J. Fréchet and Professor Michelle C. Chang

Selected Publications: • Chu, C. K.; Joseph, A. J.; Limjoco, M. D.; Yang, J.; Bose, S.; Langer, R. S.; Anderson, D. G. “Biomimetic Polymer Fiber Production from Extensible, Dynamic Hyaluronic Acid Networks in Water,” In Preparation. • Chu, C. K.; Lin, T.-P.; Shao, H.; Liberman-Martin, A.; Liu, P.; Grubbs, R. H. “Disentangling Ligand Effects on Metathesis Catalyst Activity: Experimental and Computational Studies of Ruthenium–Aminophosphine Complexes,” J. Am. Chem. Soc. 2018, 140, 5634. • Liberman-Martin, A.; Chu, C. K.; Grubbs, R. H. “Application of Bottlebrush Block Copolymers as Photonic Crystals,” Macromol. Rapid Commun. 2017, 38, 1700058. • Chu, C. K.; Ziegler, D. T.; Carr, B. M.; Wickens, Z. K.; Grubbs, R. H. “Direct Access to β-Fluorinated Aldehydes by Nitrite- Modified Wacker Oxidation,” Angew. Chem. Int. Ed. 2016, 55, 8435. • Chu, C. K.; Liang, Y.; Fu, G. C. “Silicon−Carbon Bond Formation via Nickel-Catalyzed Cross-Coupling of Silicon Nucleophiles with Unactivated Secondary and Tertiary Alkyl Electrophiles,” J. Am. Chem. Soc. 2016, 138, 6404. • Chen, M. S.; Niskala, J. R.; Unruh, D. A.; Chu, C. K.; Lee, O. P.; Fréchet, J. M. J. “Control of Polymer Packing Orientation in Thin Films through Synthetic Tailoring of Backbone Coplanarity,” Chem. Mater. 2013, 25, 4088. • Cui, L.; Cohen, J. L.; Chu, C. K.; Wich, P. R.; Kierstead, P. H.; Fréchet, J. M. J. “Conjugation Chemistry through Acetals toward a Dextran-Based Delivery System for Controlled Release of siRNA,” J. Am. Chem. Soc. 2012, 134, 15840.

Awards/Honors: Biomedical Engineering Society Career Development Award, 2019 American Chemical Society Postdoc to Faculty (ACS P2F) Workshop Selected Participant, 2019 Ruth L. Kirschstein National Research Service Award (NIH F32), NIDDK, 2018–present Koch Institute Joseph C. Jefferds, Jr. Research Travel Fellowship, 2018 Caltech Leadership Award – Commencement, 2017 AbbVie Scholars Symposium, 2016 Student Poster Competition Winner, Pacifichem, The International Chemical Congress of Pacific Basin Societies, 2015 Patricia G. Beckman Endowed Graduate Fellowship, Caltech, 2013–2014 Commencement Student Speaker, UC Berkeley College of Chemistry, 2012 Hypercube Scholar Award, UC Berkeley College of Chemistry, 2012 T. Dale Stuart Scholarship for Community Service, 2010 SIYOUNG CHOI, PhD Biomedical Engineering, Cornell University, 153 Weill Hall, Ithaca, NY, 14853 . [email protected]

Research Overview: The material properties and functions of bone extracellular matrix (ECM) are unique, as inorganic minerals deposited within organic ECM support the mechanical strength of bone and store biological molecules that regulate cellular components of bone. However, the functional role of inorganic minerals in regulating cellular behaviors and affecting bone ECM properties is not well understood. My PhD research focused on optimizing the properties of mineral coatings for application in biomedical implants. Various mineral properties influence cell fate decisions, but the conditions that would allow to control mineral formation for subsequent studies of cell behavior or clinical translation remained unclear. I developed a tunable cell culture platform with systematically controlled calcium phosphate (CaP) mineral properties. Using this enhanced throughput culture platform, I identified that optimized mineral properties enhance osteogenic differentiation of human mesenchymal stem cells (hMSC). In addition, transfection of multiple cell types using surface-mediated delivery of non-viral vectors from CaP mineral coatings can be enhanced by optimized mineral properties. As a postdoctoral and research associate, I have focused on the role of minerals in breast cancer development and progression. The specific material properties of CaP minerals found both in microcalcifications of primary breast tumors as well as in sites of bone metastasis correlate with the malignant potential of breast cancer cells and worse clinical prognosis. By systematically controlling the carbonate substitution of mineral-coated cell culture plates, I defined specific material properties of mineral that affect the malignancy of breast cancer cell. To study the effect of mineral on breast cancer bone metastasis, I adapted an in vitro biomineralization approach to enable cell culture studies on mineralized collagen, which is the basic building block of bone. Using this platform, I have found that the presence of mineral on collagen stimulates breast cancer cell adhesion and migration in a manner that is correlated with integrinmediated mechanotransduction, suggesting potential applications of this culture platform to study bone metastasis. How the changes in ECM properties that are caused by the perturbation of bone remodeling influence bone cellular components, including hMSC, immune cells and bone metastatic cancer cells, and mineral properties remain largely unknown. Future works will include the development of bone-like ECM that can be applicable to bone tissue engineering and in vitro bone disease models.

Education: - University of Wisconsin-Madison, USA Ph.D. Materials Science, May 2012/ M.S. , May 2004 - Sungkyunkwan University, KOREA M.S. Polymer Science and Engineering, February 2000/ B.S. Polymer Science and Engineering, February 1998

Research/Work Experience: - Cornell University, USA, Postdoctoral and Research Associate in Biomedical Engineering, Nov. 2012-Current ● In vitro model for bone-microenvironment effect on bone metastasis and stem cell properties of breast cancer cells/ Characterization of microcalcifications (MCs) in human breast tissues and control of hydroxyapatite for studying MCs in breast cancer - University of Wisconsin-Madison, USA, Graduate Research Assistant in Materials Science, Sep. 2004-Aug. 2012 ● Enhanced throughput screening system for mineral coated biomaterials-cell interaction/ Control of DNA delivery for multipotent stem cell transfection/ Characterization of single-stranded DNA binding and cell binding receptor density on SAM - University of Wisconsin-Madison, USA, Graduate Research Assistant in Electrical & Computer Engineering, Sep. 2002-Aug. 2004 ● Fabrication of ion channel detection system by controlling membrane size/ Stochastic resonance in ion channel system - Sungkyunkwan University, KOREA, Graduate Research Assistant in Polymer Science and Engineering, Mar. 1998-Feb. 2000 ● Characterization of thermoreversible block copolymer properties with HPLC and MALDI-TOF Mass Spectrometry

Selected Publications: - S. Choi and et al. "Intrafibrillar, bone-mimetic collagen mineralization regulates breast cancer cell adhesion and migration" Biomaterials, Vol. 198, 96-106, May 1 2018 - F. He, S. Choi* and et al. "Mineralized Cell Culture Systems for Studying Bone Metastatic Breast Cancer" In: K. Burg, ed. Engineering 3D Tissue Test Systems; Taylor & Francis. 2016 * contributed equally - S. Choi and et al. " Chemical and physical properties of carbonated HA affect breast cancer cell behavior" Acta biomaterialia, Vol. 24, 333-342, Sep. 15 2015 - B. R. Seo, P. Bhardwaj, S. Choi and et al. "Obesity-dependent changes of interstitial ECM mechanics and their role in breast tumorigenesis" Science Translational Medicine, Vol. 7, 301ra130, Aug. 19 2015 - S. Choi and et al. "Inorganic coatings for optimized non-viral transfection of stem cells" Scientific Reports, Vol. 3, 1567, Mar. 28 2013

Awards/Honors: SungKyun Honorable Scholarships, 1997 – 1999 XIAOCHUAN DAI, PhD 1Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA, 02155, 2Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139 . [email protected]

Research Overview: I propose to estabilish and lead the ‘living electronics’ research group thriving on seamless integration and functional synergy of living and non-living systems, to interrogate and direct biologically significant processes for fundentmental scientific purposes through translational territories, and eventually blur the boundaries between the livings and the non- livings.

Education: B.Sc., 2010, Peking University Ph.D., 2015, Harvard University

Research/Work Experience: My research at Harvard focued on the development of novel nano-bioelectronics platforms, with emphasis on their applications in nano-bio interfaces, electrophysiological and electromechanical studies. My research at MIT and Tufts focued on the development of functional biomaterials synergizing active biological species with nano-bioelectronics.

Selected Publications: 1. Proc. Natl. Acad. Sci. USA, 110, 6694–6699 (2013). 2. Nano Lett., 14, 1614–1619 (2014). 3. Nature Nanotechnol., 11, 776–782 (2016). 4. Acct. Chem. Res., 51, 309-318 (2018). 5. Nano Lett., 19, 2620–2626 (2019). 6. Nano Lett., doi/10.1021/acs.nanolett.9b02939 (2019)

Awards/Honors: Chinese Government Award for Outstanding Students Studying Abroad May-4th Medal, Peking University Starlight International Scholarship, Peking University National Scholarship, Peking University Samsung Scholarship, Peking University Chinese Chemistry Olympiad (CChO) Medalist Chinese Mathematics Olympiad (CMO) Medalist MATTHEW D. DAVIDSON, PhD Department of Bioengineering, University of Pennsylvania, 210 S. 33rd st, 240 Skirkanich Hall, Philadelphia, Pennsylvania, 19104 . [email protected]

Research Overview: The primary focus of my research is to understand how the structural and mechanical properties of the extracellular environment influence cell behaviors in health and disease. Specifically, I develop tunable fibrous hydrogel networks to study how changes in the mechanics of ECM networks influence cell phenotype, migration, and cell-cell interactions. My doctoral work focused on the development of high throughput in vitro disease models of fatty liver disease and fibrosis, which led to the discovery of heterotypic cell-cell interactions that promote a diseased state and combinatorial therapies to prevent diseased phenotypes. For my postdoctoral work, I combined microfabrication, multiscale mechanical characterization, and real-time imaging to develop platforms for investigating how biophysical factors contribute to the development of fibrosis. These platforms allowed for the decoupled investigation of how ECM fiber stiffness influences the differentiation of liver pericytes into myofibroblasts, and the discovery of soluble and biophysical inhibitors of activation. Additionally, I have developed collagen-inspired strain reinforcing fibrous hydrogel networks that form new mechanochemical bonds in response to physical manipulation. Mechanochemical bonds promote force induced changes in material properties (e.g. stiffness, and alignment) and can be used to fabricate macroscale structures (e.g. lumens, thick scaffolds). This work provides further explanation of how fibrotic ECM might stiffen in response to sustained cell contraction during fibrosis. In the future, my lab will develop injectable fibrous materials for engineering 3D tissue models to probe how fiber network properties influence disease progression and tissue morphogenesis as well as materials for regenerative medicine applications.

Education: • Ph.D. Biomedical Engineering, 2016, Colorado State University • B.S. Microbiology, 2012, Texas State University

Research/Work Experience: • Postdoctoral Fellow, Department of Bioengineering, University of Pennsylvania 2017-Present Advisor: Jason Burdick • Research Specialist, Department of Bioengineering, University of Illinois at Chicago, 2015-2016, Advisor: Salman Khetani • Graduate Research Assistant, Department of Biomedical Engineering, Colorado State University, 2012-2015, Advisor: Salman Khetani

Selected Publications: 1. Davidson, M.D., Ban, E, Schoonen, A.C.M., Lee, M., D’Este, M., Shenoy, V.B., Burdick, J.A. Mechanochemical Adhesion in Multi-fiber Hydrogel Networks. Submitted to Advanced Materials 2019. 2. Davidson, M.D., Song K.H., Lee, M., Llewellyn, J., Du, Y., Baker, B.M., Wells, R.G., Burdick, J.A. Engineered Fibrous Networks to Investigate the Influence of Fiber Mechanics on Myofibroblast Differentiation. ACS Biomaterials Science and Engineering, 2019, 5; 3899-3908. 3. Davidson, M.D., Kukla, D.A., Khetani, S.R. Microengineered Cultures Containing Human Hepatic Stellate Cells and Hepatocytes for Drug Development. Integrative Biology, 2017, 9; 662-677. 4. Davidson, M.D., Ballinger, K.R., Khetani, S.R. Long-term Exposure to Abnormal Glucose Levels Alters Drug Metabolism Pathways and Insulin Sensitivity in Primary Human Hepatocytes. Scientific Reports, 2016, 6; 28178. 5. Davidson, M.D., Lehrer, M., Khetani, S.R. Hormone and Drug-Mediated Modulation of Glucose Metabolism in a Microscale Model of the Human Liver. Tissue Engineering part C, 2015, 21; 716-725.

Awards/Honors: • NIH NRSA Fellowship – 2018 - Present • Center for Engineering Mechanobiology (CEMB) Teaching Award-2017 • Chicago Diabetes Day Best Poster Award - 2016 FANGYUAN DING, PhD Biology and Bioengineering, Elowitz lab, Caltech, 1200 E California Blvd, MC114-96, Pasadena, CA, 91125 [email protected]

Research Overview: I am passionate about quantitatively investigate important but previously inaccessible molecular and cellular biology questions at the single molecule level in individual mammalian cells. My interdisciplinary background allows me to integrate diverse approaches (from molecular biology to deep learning, from time-lapse microscope to mammalian cell engineering, from to integrated circuits like Arduino, etc.) to develop more sensitive and powerful single molecule tools, for probing fundamental biological questions and for biomedical applications. My work will complement conventional approaches such as genome-wide population-based studies to help generate a unified picture of RNA regulation and unlock novel RNA-based therapeutic applications.

Education: École Normale Supérieure de Paris, Ph.D. in Biophysics, Paris 2008-2012 École Normale Supérieure de Paris, Licence3 and Master. in Physics, Paris 2006-2007 Nanjing University, B.S. in Physics (rank 1st out of 221), Nanjing, China 2002-2005

Research/Work Experience: Postdoc period (Elowitz lab, CalTech): (1) Developed new image-based pipeline for quantifying single-cell RNA regulation; (2) Revealed splicing acting as a filter in an unexpected ‘economy of scale’ manner; (3) Developed label-free cell segmentation and tracking in various cell lines and imaging conditions; (4) Collaborated with Katherine Plath lab at UCLA, Mitchell Guttman lab and Changhuei Yang lab at CalTech; PhD period (Bensimon/Croquette lab, ENS Paris): (1) Developed a method to characterize the dynamics of hybridization competition using magnetic tweezers controlled by various integrated circuits; (2) Developed new ways to sequence and identify single DNA template and initiated biotech company Depixus@; (3) Collaborated with Stephen Benkovic lab at Penn State, Tom Owen-Hughes lab and Eric Le Cam lab; Master internship (Lang lab, MIT): (1) Coupled optical-tweezers with fluorescence to achieve force-fluorescence microscopy; (2) Quantified the longevity of common fluorophores when interlacing optical trap and fluorescence excitation lasers; Bachelor period (Chen Lab, NJU): Simulated the process of Metal Organic Chemical Vapor Deposition (MOCVD) of III-V semiconductors

Selected Publications: 1. Ding F, Elowitz M, Constitutive splicing and economies of scale in gene expression, Nature Structural & Molecular Biology, (2019), 26, 424. 2. Ding F, Su C, Chow KH, Elowitz M, Single cell dynamics and functions of negative autoregulatory splicing, elife (in process) 3. Ding F, et al., Displacement and dissociation of oligonucleotides during hairpin closure, Nature Chemistry (in process) 4. Ding F*, Levine JH*, Yi D, Linton J, Elowitz M, Label-free cell segmentation and tracking in various cell lines and imaging conditions by deep learning (in preparation) 5. Ding F, Manosas M, Spiering MM, Benkovic SJ, Bensimon D, Allemand JF, Croquette V, Single-molecule mecha-nical identification and sequencing, Nature Methods, (2012) 9(4), 367-372. (News and Views: S Linnarsson, Nature Methods) 6. Ferrer J*, Ding F*, Tarsa P, Brau R, Lang M, Interlaced Optical Trapping and Fluorescence Excitation improves fluorophore longevity, Curr. Pharm. Biotechnol. (2009) (5): 502-7. 7. Ding F, Pan L, Yu Y, Su W, Discussion and modification of experiment of measuring the heat conduction of coefficient of gas based on the hot-wire method, Physics Experimentation (2004) 24(12):39-41-7.

PATENTS licensed by Depixus@ 1.Bensimon D, Croquette V, Allemand JF, Manosas M, Ding F, Method of DNA sequencing by polymerization, EP2390350A1, WO2011147929A1, US20130171636A1, et al.; 2.Bensimon D, Allemand JF, Manosas M, Ding F, Croquette V, Method of DNA sequencing by hybridization, EP2390351A1, WO2011147931A1,US20130137098A1, et al.; 3.Bensimon D, Allemand JF, Ding F, Croquette V, Method of DNA detection and quantification by single-molecule hybridization and manipulation,WO2013093005A1, EP2794915A1, US20150031028A1, et al. 4.Bensmion D, Croquette V, Gouet G, Allemand JF, Ding F, Process for detection of DNA modifications and protein binding by single molecule manipulation, CA2898151A1, WO2014114687A1.

Awards/Honors: 2018-2021 IST/AWS Cloud Credit Program; 2018-2019 Intel corporation research funding 2013-2015 Schlumberger Faculty for the Future fellowship 2005-2012 Fellowship at École Normale Supérieure de Paris (10 international students worldwide) 2002-2005 1st Rank Outstanding Student Scholarship; 2002 Samsung Scholarship, Nanjing University (1 out of 220 students) Y. DING, PhD 1Medicine, UCLA, 650 Charles E Young Dr S, Los Angeles, California, 90095, 2Bioengineering, UCLA, 650 Charles E Young Dr S, Los Angeles, California, 90095 . [email protected]

Research Overview: My passion for a career in science has always motivated me to pursue a broad field of knowledge and experience throughout my academic training. During my undergraduate study at Tsinghua University in China, I became fascinated with the application of methods to solve issues in the biomedical / biomechanical fields, which laid a foundation for my PhD in the biomedical optics at Peking University. The central theme of my scientific training in China has been to develop novel photonic systems for biomedical and translational research. To further translate my research to advance the field of developmental cardiac mechanics, I am pursuing multi-disciplinary research by establishing a multi- scale imaging strategy along with an advanced computational framework. Under the supervision of Dr. Tzung Hsiai at UCLA, I collaborate with my colleague to develop a non-axially-scanned sub-voxel resolution LSFM (SV- LSFM), allowing for high-throughput volumetric imaging at the subcellular level in an extended field-of-view. I integrated this imaging platform with the displacement analysis of myocardial mechanical deformation (DIAMOND) developed by me and my colleagues to quantify doxorubicin-induced myocardial injury and regeneration (AHA CDA). I have also collaborated with biomedical researchers and physician-scientists across the US in multiple projects, including in vivo tracking of neural crest cell migration into the developing heart tube (NIH K99), capturing 4-D dynamics of atrio- ventricular valve leaflets throughout the cardiac cycle in zebrafish, tracing stem cell lineage, localizing 3-D vascular calcification, and investigating developmental pulmonary mesenchyme in mouse models. To extend my research on the comprehensive exploration of advanced imaging and computational methods, I challenged myself to establish the first framework by integrating LSFM with virtual reality (VR) for an interactive and immersive microenvironment in which we recapitulate developmental cardiac mechanics and physiology with high spatiotemporal resolution. To enhance efficiency and accuracy of myocardial segmentation for 4-D DIAMOND analysis and VR-LSFM, I have adapted the subspace approximation with augmented kernels (Saak) transform-based machine learning for cardiac microscopy images. Collectively, this multi-disciplinary training has prepared me with the necessary knowledge and expertise to pursue an independent academic career. I will continue integrating advanced imaging with machine learning and interactive quantification to advance developmental cardiac mechanics.

Education: 2010-2015 Ph.D., Biomedical Engineering Peking University, Beijing, China 2006-2010 B.S., Mechanical Engineering Tsinghua University, Beijing, China

Research/Work Experience: 2017-Present Assistant Project Scientist UCLA School of Medicine, Los Angeles, CA 2015-2017 Visiting Assistant Project Scientist UCLA School of Medicine, Los Angeles, CA

Selected Publications: 1) Y. Ding†, A. Abiri†, et al., JCI Insight, 2(22): e97180, 2017. 2) J. Chen†, Y. Ding†, et al., JCI Insight, 4(8), e125362, 2019. 3) P. Fei†, J. Nie†, J. Lee†, Y. Ding†, et al., Advanced Photonics, 1(1): 016002, 2019. 4) Y. Ding†, M. Zhang†, et al., Journal of Biophotonics, 8(1-2), 94-101, 2015. 5) Y. Ding†, H. Xie†, et al., Optics Express, 20(13), 14100-14108, 2012.

Awards/Honors: 1) 2019-2024 NIH K99HL148493 2) 2018-2021 AHA 18CDA34110338 3) 2013-2014 1st Award for 13th National "Challenge Cup" Academic Science and Technology Competition, China MEGHAN C. FARRELL-FAIRBANKS, PhD Integrated Mathematical Oncology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, SRB4, Tampa, FL, 33612 [email protected]

Research Overview: My doctoral work focused on a mechanistic understanding of the cysteine cathepsin proteolytic network dynamics and its role in tissue remodeling. My postdoctoral research in mathematical oncology shifted my focus to using systems biology approaches to cultivate a mechanistic understanding of cancer heterogeneity and evolution to optimize treatment strategies. One project investigates the underlying evolutionary dynamics of clonal hematopoietic diseases such as myelodysplastic syndromes and chronic myelomonocytic leukemia. To date, my efforts have focused on quantifying heterogeneity in leukemia samples, compared to healthy cases, in attempts to identify key phenotypic signatures that can ultimately be used prognostically in the clinical setting. Specifically, I have developed a pipeline for analyzing single cell RNA-sequencing data and establishing that a generalized diversity score can successfully discriminate between normal and malignant tissues (work published earlier this year in JCO Clinical Cancer Informatics). The other research projects of my postdoctoral work revolve around mechanistic mathematical modeling of open questions in cancer research. One project involves modeling of cell-to-cell interactions in a lymphoma-fibroblast-macrophage environment, to evaluate the role of a third player, lymphoma, to otherwise homeostatic interactions between fibroblast and macrophages. My most recent project focuses on the emerging field of cardio-oncology and using mathematical modeling to determine patient-specific accumulation of amyloid proteins in the heart leading to heart failure in multiple myeloma patients.

Education: PhD, Biomedical Engineering, 12/2017, Georgia Tech & Emory University BS, Mechanical Engineering, 5/2012, University of Florida

Research/Work Experience: • Postdoctoral Fellow, Moffitt Cancer Center and Research Institute, Integrated Mathematical Oncology (2017-Present), PI: Philipp M. Altrock, PhD • Graduate Research Assistant, Georgia Institute of Technology, Biomedical Engineering (2017-2012), PI: Manu O. Platt, PhD • Biomedical Informatics/SPARK Research Intern, Moffitt Cancer Center and Research Institute, Biomedical Informatics (Summers 2010, 2011, 2012), PI: Steven Eschrich, PhD • Undergraduate Researcher, University of Florida, Space, Automation and Manufacturing Mechanisms Lab (2010-2012), PI: Gloria Wiens, PhD

Selected Publications: • Ferrall-Fairbanks MC, Barry ZT, Affer M, Shuler MA, Moomaw EW, Platt MO. “PACMANS: A Bioinformatically Informed Algorithm to Predict, Design, and Disrupt Protease-on-Protease Hydrolysis.” Protein Science, 2017 Apr; 26(4):880-890. doi: 10.1002/pro.3113 PMID: 28078782 • Cvetkovic C*, Ferrall-Fairbanks MC*, Ko E, Grant L, Kong H, Platt MO, Bahir R. “Investigating the Life Expectancy and Proteolytic Degradation of Engineered Skeletal Muscle Biological Machines.” (* co-first author) Scientific Reports, 2017 Jun 19;7(1):3775. doi: 10.1038/s41598-017-03723-8 PMID: 28630410 • Ferrall-Fairbanks MC*, West DM*, Douglas SA, Averett RD, Platt MO. “Computational predictions of novel cysteine cathepsinsmediated fibrinogen degradation.” (* co-first author) Protein Science, 2018 Mar; 27(3):714-724. doi: 10.1002/pro.3366 PMID: 2926658 • Grant L*, Raman R*, Cvetkovic C, Ferrall-Fairbanks MC, Diaz GP, Hadley P, Platt MO, and Bashir R. “Long-term cryopreservation and revival of tissue engineered skeletal muscle.” (* co-first author) Tissue Engineering Part A. 2019 Jan 9. doi: 10.1089/ten.TEA.2018.0202 PMID: 30412045 • Ferrall-Fairbanks MC, Ball M, Padron E, Altrock PM. “Leveraging single cell RNA sequencing experiments to model intra- tumor heterogeneity.” JCO Clinical Cancer Informatics. 2019 Apr: 3:1-10. doi: 10.1200/CCI.1800074 PMID: 30995123

Awards/Honors: 2019, 2018 - BMES Career Development Award 2018 - Moffitt's Junior Scientist Partnership Award 2015 - Georgia Tech Faces of Inclusive Excellence 2013 - Tau Beta Pi Fellow 2013 - NSF GRFP Honorable Mention VINCENT F. FIORE, PhD Laboratory for Mammalian Cell and , Rockefeller University [email protected] Research Overview: Tissues are precisely built in their three-dimensional shape and the arrangement of specific cell types. How this architecture emerges – through biophysical forces and gene regulatory networks that must coordinate across length scales – is a fundamental question in biology and tissue engineering. My global objective is to understand how complex tissue architectures emerge during development and how they go array in disease. This work is stimulated by doctoral and postdoctoral research, where I’ve uncovered novel mechanisms of how cells sense and respond to mechanical forces across diverse biological length scales. At the single-molecule level, I discovered how dual binding of an extracellular ligand by integrin and a co-receptor caused force-triggered affinity, elucidating a novel mechanochemical molecular switch4. At the cell level, I discovered that coupling between the on-rate of integrin-ECM bonds and the recruitment of signaling effectors tunes how cells sense ECM stiffness1,3. In my postdoctoral work at the tissue scale, by combining computational predictions with biophysical measurements and in vivo genetic manipulations, I’ve uncovered that the mechanical forces generated by differentiated cells and the ECM encodes the diverse tumor architectures seen within complex epithelia (manuscript in review). Importantly, the dynamic interplay of these forces coalesces at the epithelial- stromal boundary to govern the invasive and cell division behaviors of cancer stem cells. In the future, I aim to develop a systems-level understanding of how forces are transduced and integrated across length scales to specify cell fates and shape complex tissues. I will explore the broad hypothesis that distinct cell types are tuned to sense and respond to ‘niche-specific’ mechanical forces, which ultimately dictates their cell-type-specific biological function within a tissue. Particular focus will be paid to the assembly, molecular diversity, and material properties of the ECM, which my work has shown has a major role in sculpting tissue architectures and specifying cell fates during tumorigenesis. I will accomplish this by 1) developing a platform of mouse genetic tools to manipulate forces, at will, on specific cell types in vivo, and 2) integrating genomic tools to comprehensively understand the cell- and tissue-level biological consequences with molecular resolution. Altogether, my work will propel an integrated understanding of how forces shape tissue architecture, from molecular to tissue scales in living organisms.

Education: • 2014, Ph.D. Biomedical Engineering, Georgia Institute of Technology and Emory University • 2008, B.S.E. Biomedical Engineering, Pennsylvania State University

Research/Work Experience: • 2015-present, Postdoctoral Fellow, The Rockefeller University • 2008-2014, Graduate Research Fellow, Georgia Institute of Technology and Emory University • 2006-2008, Research and Development Intern, NanoHorizons • 2006-2008, Undergraduate Research Assistant, Pennsylvania State University

Selected Publications: 1. Fiore VF, Wong S, Tran C, Tan C, Xu W, Sulchek T, White ES, Hagood JS, Barker TH. Integrin αvβ3 drives fibroblast contraction and strain stiffening of soft provisional matrix during progressive fibrosis. JCI Insight. 3(20), 2018. 2. Ouspenskaia T, Maros I, Mertz AF, Fiore VF, Fuchs E. Wnt-SHH antagonism specifies and expands stem cells prior to niche formation. Cell. 164(1-2):156-69, 2016. (*Highlighted in Nature Reviews Molecular ) 3. Fiore VF, Strane PW, Bryksin AV, White ES, Hagood JS, Barker TH. Conformational coupling of integrin and Thy-1 regulates Fyn priming and fibroblast mechanotransduction. J Cell Biol. 211:173-90, 2015. (*Highlighted as In Focus for Journal of Cell Biology) 4. Fiore VF*, Ju L*, Chen Y, Zhu C, Barker TH. Dynamic catch of a 5 1-Thy-1-syndecan-4 trimolecular complex. Nature Commun. 5:4886, 2014. (*Equal contribution) 5. Fiore VF, Lofton MC, Roser S, Yang SC, Roman JR, Murthy N, Barker TH. Polyketals: A new biocompatible polymer for lung therapeutics delivery. Biomaterials 31(5): 810-817, 2010.

Awards/Honors (selected): • 2018, Charles H. Revson Senior Postdoctoral Fellowship (*1 of 6 in Tri-state area) • 2018, American Society for Matrix Biology Founders Award (*one award given internationally) • 2015, Ruth L. Kirschstein Institutional National Research Service Award (T32) • 2015, Best Fundamental Research Award, Georgia Tech Department of Biomedical Engineering • 2014, Sigma Xi Best Ph.D. Thesis Award • 2010, National Science Foundation Graduate Research Fellowship ANOOSHA FORGHANI, PhD Biomedical Engineering, Penn state university, N 023 Millennium Science Complex, University Park, PA, 16802 [email protected]

Research Overview: - Fabrication of quasi-3D vascularized bone like constructs and characterizing the constructs both in vitro and in vivo. - Endothelial differentiation using adipose tissue derived CD34+/CD31- progenitors. - Spatiotemporal release of miRNAs using gold and silver nanoparticles. Education: PhD in Biomedical Engineering , 2019 , Pennsylvania state University. M.S. in Mechanical Engineering, 2016, Louisiana State University. B.S.in Materials Science and Engineering , 2011 , Isfahan University of Technology. Research/Work Experience: Penn State University -Study endothelial differentiation of Adipose Derived progenitors. -Develop a quasi 3D vascularized bone like construct. -Study osteogenesis differentiation of Bone Marrow Stem Cells using heterodimer Gold Iron Oxide nanoparticles for deep tissue regeneration applications. -Regulate dual gene by Gold and Silver nanoparticles with different plasmonic properties. Louisiana State University -Develop an injectable Thiol-Acrylate based polymer for bone tissue engineering applications. -Fabricate cell Sheets using thermally reversible hydrogels. Isfahan University of Technology -Evaluate bioactivity and biocompatibility of Forsterite-Fluorapatite composite nanopowder for biomedical applications. -Fabricate Forsterite nanofibers using Electrospinning Technique. Selected Publications: Anoosha Forghani, Mahta Mapar, Mahshid Kharaziha, Mohammad H. Fathi, “Fabrication and characterization of novel nanocomposite powder for oral bone defects”, International Journal of Applied Ceramic Technology,Vol (10), E282-E289 ,2013. S.M.Mirhadi, A.Forghani, F.Tavangarian, “A modified method to synthesize single-phase forsterite nanoparticles at low temperature”, Ceramics International, Vol (42), 7974-7979, 2016. Forghani A, Kriegh L, Hogan K, Chen C, Brewer G, Tighe TB, Devireddy R, Hayes D “Fabrication and characterization of cell sheets using Methylcellulose and PNIPAAm thermoresponsive polymers: A comparison study”, Journal of Biomedical Materials Research Part A 105 (5), 1346-1354,2017. Anoosha Forghani, Ram Devireddy, “Methylcellulose Based Thermally Reversible Hydrogels”, Springer, Adipose-Derived Stem Cells: Methods and Protocols, 41-51, 2018. Anoosha Forghani, Leah Garber, Cong Chen, Fariborz Tavangarian, Timothy B. Tighe, Ram Devireddy, John A. Pojman, Daniel Hayes, “Fabrication and characterization of Thiol- Triacrylate polymer via Michael Addition Reaction for biomedical applications”, Biomedical Materials, 2018. Fariborz Tavangarian, Abbas Fahami, Guoqiang Li, Mohammadhassan Kazemi, Anoosha Forghani, “Structural characterization and strengthening mechanism for forsterite nanostructured scaffolds synthesized by multistep sintering method”, Journal of Materials Science and Technology, 34 (12), 2263-2270, 2018. Shue Li, John Nicholas Poche, Yiming Liu, Thomas Scherr, Jacob McCann, Anoosha Forghani, Mollie Smoak, Mitchell Muir, Cong Chen, Dino J. Ravnic, Daniel Hayes, “Hybrid Synthetic‐Biological Hydrogel System for Adipose Tissue Regeneration”, Macromolecular Bioscience, DOI: 10.1002/mabi.201800122, 2018. Fariborz Tavangarian, Caleb A Zolko, Abbas Fahami, Anoosha Forghani, Daniel Hayes, “Facile Synthesis and Structural Insight of Nanostructure Akermanite Powder”, Ceramics International, 2019. Srinivas V. Koduru,, Ashley N. Leberfinger, Denis Pasic, Anoosha Forghani, Shane Lince, Daniel J. Hayes, Ibrahim T. Ozbolat, Dino J. Ravnic, “Cellular Based Strategies for Microvascular Engineering”, Stem Cell Reviews and Reports, (2019) 15:218–240 . Anoosha Forghani, Srinivas Koduru ,Cong Chen, Ashley Leberfinger, Dino Ravnic, Daniel Hayes, “Differentiation of Adipose Tissue Derived CD34+/CD31- Cells into Endothelial Cells in Vitro”, Regenerative Engineering and Translational Medicine ,doi.org/10.1007/s40883-019-00093-7. Anoosha Forghani, Srinivas V. Koduru, Dino J. Ravnic, Daniel Hayes"Endotheliogenic/Osteogenic co-differentiation of multipotent progenitors in quasi-3D cell sheet stacks for vascularized bone regeneration", Biomaterials , Submitted. Awards/Honors: -Enrichment Award, Louisiana State University, 2013 -Invited speaker for Materials Research Institute (MRI) facility at Penn State University (October 2017). -Reviewed papers for: Journal of Biomedical Materials Research: Part A (2018), Journal of Materials Science: Materials in Medicine (2018), Sensors & Actuators: B. Chemical (2017), Materials Science & Engineering A (2015), IEEE Transactions on NanoBioscience (2015), Journal of Biomedical Materials Research: Part A (2014), Journal of Alloys and Compounds (2014) FARZAD FOROUZANDEH, PhD Microsystem Engineering, Rochester Institute of Technology, 168 LOMB MEMORIAL DR BLDG 17, ROCHESTER, NEW YORK, 14623 . [email protected] Research Overview: 1. Biomedical Microsystems 2. Drug delivery Microdevices 3. Polymer MEMS 4. Microscale sensors and actuators 5. Microscale 3D-printing technologies 6. Bio-Microfluidics

Education: 1. PhD in Microsystems Engineering (2019) Rochester Institute of Technology (RIT), Rochester, NY, USA Advisor: Dr. David Borkholder Thesis: Implantable Microsystem Technologies For Nanoliter-Resolution Inner Ear Drug Delivery 2. M.Sc. in Biomedical Engineering (2013) Tehran Polytechnic (Amirkabir University of Technology), Tehran, Iran GPA: 3.91 3. B.Sc. in Mechanical Engineering (2010) Tehran Polytechnic (Amirkabir University of Technology), Tehran, Iran

Research/Work Experience: 1. Co-managing a R01 NIH grant to develop an implantable microsystem for inner ear drug delivery 2. Co-managing collaboration with surgeons for implantable product optimization and in vivo animal experiments 3. Co-writer of a R01 NIH grant 4. Directly managed 3 PhD students, 2 MSc student, 3 undergraduate student, 2 student workers, and multiple vendors. 5. Design, fabrication, test, characterization, and packaging of biomedical microdevices 6. 3D-printing technology development (inkjet, SLA, Aerosol, photonic sintering, etc.) 7. MEMS fabrication technology (CVD (parylene-C), photolithography) 8. Conventional fabrication methods (Machining, CNC, Laser cutter table) 9. CAD modeling & Numerical simulation of biomedical devices (expert level knowledge of COMSOL and SOLIDWORKS) 10. Microsystem evaluation methods (SEM, confocal and optical microscope, profilometer)

Selected Publications: 1. Forouzandeh F., Zhu X., Alfadhel A., Ding B., Walton J.P., Cormier D., Frisina R.D., Borkholder D.A., 2018. A nanoliter resolution implantable micropump for murine inner ear drug delivery. Journal of controlled release, 298, pp.27-37. 2. Forouzandeh F., Zhu X., Alfadhel A., Ding B., Walton J.P., Cormier D., Frisina R.D., Borkholder D.A., 2018. A wirelessly controlled fully implantable microsystem for nano-liter resolution inner ear drug delivery. Proc. 18th Solid- State Sens., Actuators, Microsyst. Workshop, Hilton Head Island, SC, USA, pp. 38–41. (Oral presentation) 3. Hsu, M.C., Alfadhel, A., Forouzandeh, F. and Borkholder, D.A., 2018. Biocompatible magnetic nanocomposite microcapsules as microfluidic one-way diffusion blocking valves with ultra-low opening pressure. Materials & Design, 150, pp.86-93. 4. Alfadhel, A., Ouyang, J., Mahajan, C.G., Forouzandeh, F., Cormier, D. and Borkholder, D.A., 2018. Inkjet printed polyethylene glycol as a fugitive ink for the fabrication of flexible microfluidic systems. Materials & Design, 150, pp.182-187.

Awards/Honors: • Outstanding scholarship invited presenter in RIT graduate showcase, fall 2017 • Ranked 2nd of GPA in M.Sc. in the biomechanical group of Biomedical Eng. Dep., 2012 • Ranked 171th in the nationwide university exam for M.Sc. in Mechanical Engineering(among more than 19000 participants),2010 • Ranked 316th in the nationwide university entrance exam, Mathematics and Physics Discipline (among more than 400,000 participants), 2005 • Admitted to semi-final of Iranian Physics Olympiads, 2003 BENJAMIN R. FREEDMAN, PhD Wyss Institute for Biologically Inspired Engineering at Harvard University, 58 Oxford St, Cambridge, Massachusetts, www.benjaminrfreedman.com . [email protected]

Research Overview: My research focuses on the design and synthesis of biomaterials to augment biological tissues, with a special focus in orthopaedics. These biomaterials are designed to provide an adhesive interface to anchor directly to tissue surfaces and control release of therapeutics to improve tissue healing. During my time in graduate school I used many novel in vivo animal models, mechanical testing methods, imaging modalities, and microscopy techniques to study the role of healing, proteoglycans, collagen type V, and fatigue loading on tendon. I realized that knowledge of biomaterials and therapeutic delivery strategies would be essential to develop treatments for tendon. As an NRSA F32 postdoctoral fellow, I have applied my knowledge of tendons and the extracellular matrix in healthy and pathological states to develop novel biomaterials. During my postdoc, we have developed and explored the capacity of tough adhesive biomaterials, inspired by the mucus secreted by slugs, to adhere strongly to tendon surfaces. The goal of this tough adhesive biomaterial is to provide mechanical support and a template for tendon regeneration, serve as a depot for local delivery of agents, support cell growth and infiltration, and provide gliding of surrounding tissues. My future research aims to develop a new cell delivery strategy to dynamically recruit cells in vivo, expand them, and release them on-demand to promote tendon healing throughout aging using this adhesive biomaterial platform (NIH K99/R00 in review).

Education: 2017- Postdoctoral Fellow, Wyss Institute for Biologically Inspired Engineering at Harvard University 2017 Ph.D., Bioengineering, University of Pennsylvania, Philadelphia, PA 2011 B.S., Biomedical Engineering, University of Rochester, Rochester, NY

Research/Work Experience: 2017- Postdoctoral Fellowship (NIH, NIA, NRSA F32): On-Demand Stem Cell Delivery Systems for Tendon Healing Throughout Aging. Advisor: David J Mooney 2011-2017: Graduate Research: (NIH T32, NSF GRFP): Multiscale Mechanical, Structural, and Compositional Response of Tendon to Dynamic Loading During Healing. Advisor: Louis J Soslowsky 2014-2015: Penn Center for Innovation Technology Transfer Fellow, University of Pennsylvania, Philadelphia, PA. 2011: Intramural Research Training Award (IRTA), National Institutes of Health, Bethesda, MD. Advisor: Fran Sheehan 2010: Bioengineering Summer Internship Program (BESIP), National Institutes of Health, Bethesda, MD. Advisor: Fran Sheehan

Selected Publications: 1. Freedman BR, Mooney DJ. Biomaterials to Mimic and Heal Soft Connective Tissues. Advanced Materials, February 2019. [ePub] 2. Blacklow SA, Li J, Freedman BR, Chen C, Mooney DJ. Bioinspired, Antimicrobial, Biologic-Free, Biomechanically Active Hydrogel Dressings for Accelerating Wound Healing. Science Advances, May 2019. 3. Freedman BR, Rodriguez AB, Leiphart R, Newton J, Ban E, Sarver J, Mauck RL, Shenoy VB, Soslowsky LJ. Dynamic Loading and Tendon Healing Affect Multiscale Properties and ECM Stress Transmission. Nature Scientific Reports, June 2018. 4. Freedman BR, Rodriguez AB, Hillin CD, Weiss SA, Han B, Han L, Soslowsky LJ. Tendon Healing Affects the Multiscale Mechanical, Structural, and Compositional Response to Quasi-Static Loading. J Royal Society Interface, January 2018. 5. Freedman BR, Salka NS, Morris TM, Nuss CA, Riggin CN, Gordon JA, Shen A, Farber DC, Soslowsky LJ. Temporal Healing of Achilles Tendons Following Injury Depends on Surgical Treatment and Return to Activity Timing. JAAOS, February 2017. 6. Freedman BR, Zuskov A, Sarver JJ, Buckley MR, Soslowsky LJ. Evaluating Changes in Tendon Crimp With Fatigue Loading as an Ex Vivo Structural Assessment of Tendon Damage. J Orthopaedic Research, April 2015.

Awards/Honors: ·Ruth L. Kirchstein National Research Service Award (NRSA, F32, NIA, NIH), 2017. ·NIH/NIAMS Training in Musculoskeletal Research T32 Trainee, 2015-17. ·NSF Graduate Research Fellowship, 2011. ·New Investigator Research Award (NIRA) Winner, ORS 2019. ·Solomon Pollack Award for Excellence in Bioengineering Graduate Research, UPenn, 2017. ·President Gutmann Leadership Award, University of Pennsylvania, 2016. ·Visiting Junior Scientist (HHMI), University of Puerto Rico Mayagüez, 2016. RUIXUAN GAO, PhD 1McGovern Institute for Brain Research and Media Lab, MIT, 43 Vassar St., Building 46-2171B, Cambridge, MA, 02139, 2HHMI . Janelia Research Campus, 19700 Helix Dr, Ashburn, VA, 20147 . [email protected]

Research Overview: The human brain contains 80 billion neurons interconnected through ~7,000 synapses per each of them: each of these synapses includes tens of thousands of molecules that work concertedly to regulate synaptic transmission. Understanding such a complex system at molecular level is one of the most audacious challenges in science and technology today. My long-term research goal is to develop methods to capture the architecture, function, and dynamics of cells and tissues with molecular contrast, nanoscale resolution, and sub-ms temporal resolution. Toward this goal, I propose to design optical, biomolecular, and chemical tools for (1) in-situ superresolution imaging of endogenous biomolecules (proteins, RNAs, lipids, etc.) in cells and intact tissues with single-molecule sensitivity, (2) high-throughput mapping of cellular connections (connectome) and molecular distributions (transcriptome, proteome, etc.) across the entirety of an organ, such as the brain, (3) molecularly resolved tracking of misfolded proteins and their pathological effects, and (4) perturbation and recording of cellular dynamics via multi-photon excitation and nanoelectronic detection for multiplexed brain- machine interface.

Education: •Ph.D. in Physical Chemistry (2015), Harvard University •B.S. in Chemistry (2009), University of California at Berkeley

Research/Work Experience: •Nanoscale Imaging of cells and tissues Postdoctoral Associate (PI: Ed Boyden), MIT, Oct. 2015 – present Visiting Postdoctoral Associate (PI: Eric Betzig), HHMI, Janelia Research Campus, Jan. 2016 – present •Design, Fabrication, and Characterization of Nanoelectronic Devices for Nano-Bio Interface Graduate Research Assistant (PI: Charles Lieber), Harvard University, Dec. 2009 – June 2015

Selected Publications: •R. Gao*, C.-C. Yu*, L. Gao, Y.-Y. Chou, S. Upadhyayula, E.S. Boyden, “A structurally homogenous polymer network for physical expansion of biological specimens,” Manuscript in preparation (2019). •R. Gao*, S.M. Asano*, S. Upadhyayula*, I. Pisarev, D.E. Milkie, T.-L. Liu, V. Singh, A. Graves, G.H. Huynh, Y. Zhao, J. Bogovic, J. Colonell, C.M. Ott, C. Zugates, S. Tappan, A. Rodriguez, K.R. Mosaliganti, S.G. Megason, J. Lippincott- Schwartz, A. Hantman, G.M. Rubin, T. Kirchhausen, S. Saalfeld, Y. Aso, E.S. Boyden, E. Betzig, “Cortical column and whole-brain imaging with molecular contrast and nanoscale resolution,” Science 363, eaau8302 (2019). •D. Oran*, S.G. Rodriques*, R. Gao, S.M. Asano, M.A. Skylar-Scott, F. Chen, P.W. Tillberg, A.H., Marblestone†, E.S. Boyden†, “3-D nanofabrication by volumetric deposition and controlled shrinkage of patterned scaffolds,” Science 362, 1281-1285 (2018). •S.M. Asano*, R. Gao*, A.T. Wassie*, P.W. Tillberg, F. Chen, E.S. Boyden, “Expansion microscopy: protocols for imaging proteins and RNA in cells and tissues,” Curr. Protoc. Cell Biol., e56 (2018). •R. Gao*, S.M. Asano*, E.S. Boyden, “Q&A: Expansion microscopy”, BMC Biology 15, 50 (2017). •9. R. Gao*, I. Gupta*, E.S. Boyden, “Sonofragmentation of ultrathin 1D nanomaterials”, Part. Part. Syst. Charact. 34, 1600339 (2017). •Y-S. No*, R. Gao*, M. Mankin, R. Day, H-G. Park, C.M. Lieber, “Encoding active device elements at nanowire tips,” Nano Lett. 16, 4713–4719 (2016). •R. Gao, S. Strehle, B. Tian, T. Cohen-Karni, P. Xie, X. Duan, Q. Qing, C.M. Lieber, “Outside looking in: Nanotube transistor intracellular sensors,” Nano Lett. 12, 3329-3333 (2012). •X. Duan, R. Gao, P. Xie, T. Cohen-Karni, Q. Qing, H-S. Choe, B. Tian, X. Jiang, C.M. Lieber, “Intracellular recordings of action potentials by an extracellular nanoscale field-effect transistor,” Nature Nanotechnol. 7, 174-179 (2012).

Awards/Honors: •Dudley R. Herschbach Teaching Award (Department of Chemistry and Chemical Biology, Harvard University, 2015) •Distinction in Teaching Awards (Harvard University, 2011 – 2013) •Japan Student Services Organization (JASSO) Graduate Fellowship (one of the few selected for full scholarship, 2009 – 2014) BRYAN C. GOOD, PhD Department of Biomedical Engineering, Pennsylvania State University, 122 Chemical and Biomedical Engineering Building, University Park, PA, 16802, [email protected]

Research Overview: My long-term research interests involve the improvement of clinical therapies for cardiovascular disease. I am passionate about working in collaboration with clinicians to identify the problems that they encounter on a daily basis. Additionally, I feel very strongly about the need to combine computational modeling with extensive experimental validation. While computational work continues to become more prominent in our field, complex simulations are often performed with little to no experimental validation and significantly hurt the overall impact of the work. A majority of my research experience has revolved around studying medical devices for patients with cardiovascular disease. While my primary focus during my PhD was on the Penn State Pediatric Ventricular Assist Device, I also studied the Thoratec HeartMate II and FDA benchmark pumps, and as part of my postdoctoral training have been developing a centrifugal pump for Fontan patients. These projects have allowed me to study both the fluid dynamics using laser-based flow visualization and also the biocompatibility of the blood pump materials using blood handling and staining techniques and confocal and scanning electron microscopy. Most recently, I have been working directly with a clinical collaborator at the Penn State Neuroscience Institute on improving surgical therapies for acute ischemic stroke patients. Through this project, I have gained valuable experience shadowing in the operating room and endovascular suite and spending time in the clinic observing patients before and after surgery. Through both experimental benchtop studies using ex vivo patient samples and computational modeling, our goal is to expand our understanding of how blood clots lodge in cerebral arteries and improve current catheter based surgical devices and techniques.

Education: • Pennsylvania State University, May 20107, PhD in Bioengineering • Carnegie Mellon University, May 2012, BS in Mechanical Engineering and BS in Biomedical Engineering

Research/Work Experience: • American Heart Association Postdoctoral Fellow, Hershey Neuroscience Institute and Penn State University, January 2019-present, Advisors: Dr. Scott Simon, Dr. Keefe Manning, Dr. Francesco Costanzo • Postdoctoral Research Scholar, Penn State University, 2017-2018, Advisors: Dr. William Weiss and Dr. Keefe Manning • Graduate Research Assistant, Penn State University, 2012-2017, Advisor: Dr. Keefe Manning

Selected Publications: 1. Good B, Simon S, Manning K, Costanzo F. Development of a Computational Model for Investigating Cyclic Aspiration on Thromboemboli Removal in Acute Ischemic Stroke. In Review at BMMB. 2. Hariharan P, Aycock A, Buesen M, Day S, Good B, et al. Interlaboratory Particle Image Velocimetry measurement of velocity field in the FDA blood pump geometry. CVET. 1-18. 2018. 3. Good B, Manning K. Asynchronous Pumping of a Pulsatile Ventricular Assist Device in a Pediatric Anastomosis Model. World Journal for Pediatric and Congenital Heart Surgery. World J Pediatr Congenit Heart Surg. 8(4):511-519. 2017. 4. Good B, Deutsch S, and Manning KB. Continuous and Pulsatile Pediatric Ventricular Assist Device Hemodynamics with a Viscoelastic Blood Model. CVET. 7(1):23-43. 2016. 5. Good B, Deutsch S, and Manning KB. Hemodynamics in a Pediatric Ascending Aorta Using a Viscoelastic Pediatric Blood Model. Annals of biomedical engineering. 1-17. 2015.

Awards/Honors: • Trainee Scholarship, Michigan Cardiovascular Innovation and Translation Workshop, 2017 • Koff Award for Outstanding Abstract, ASAIO 2016 Conference • Villforth PSU Graduate Award, Spring 2016 • Riess PSU Graduate Award, Spring 2015 • International Society of Biorheology Travel Award, 2015 JOSEPH CLARK GRIM, PhD 1Department of Chemical and Biological Engineering, University of Colorado-Boulder, 3415 Colorado Ave., 596 UCB, Boulder, CO, 80303, 2BioFrontiers Institute, Boulder, CO, 80303 . [email protected]

Research Overview: The long-term goals of my research program are to elucidate how innate immunity influences the progression of fibrotic diseases and to develop strategies to treat fibrosis through immunomodulation. My research will be highly interdisciplinary, relying on fundamental\ advances in organic chemistry and materials science enabling the design of responsive hydrogel culture platforms that mimic tissue dynamics to model fibrosis in vitro. Using these platforms, I seek to identify new mechanisms of innate immunological memory and explore how this memory drives the progression of pathological fibrosis. Leveraging these findings, I will also engineer materials that temper the immune response in vivo as a generalizable strategy to treat fibrosis. Initially, my lab will focus on the following research areas: 1. Design photo-responive hydrogels to identify epigenetic mechanisms of mechanical memory in innate immune cells and their contribution toward fibrosis 2. Develop nanomaterials that target to bone marrow progenitors to inhibit trained immunity as a strategy to treat fibrosis 3. Engineer immunomodulatory polymers that target to fibrotic tissues and dampen the immune response using chemical biologybased agonists of immune cell receptors

Education: Ph.D. Organic Chemistry, University of Wisconsin-Madison, 2014 B.S. Chemistry, University of Wisconsin-Green Bay, 2008

Research/Work Experience: Postdoctoral Research Fellow (Advisor: Kristi Anseth), University of Colorado-Boulder (2014 – Present) My postdoctoral research focused on the development of new photochemical reactions that enable reversible control of the mechanical and biochemical properties of hydrogels. I employed these materials to understand the role that macrophages and mechanical memory play in mediating cardiac fibrosis. Graduate Research Fellow (Advisor: Laura Kiessling), University of Wisconsin-Madison (2008 – 2014) In my graduate research, I designed small molecule and polymeric probes of C-type lectin receptors on immune cells. Using these tools, I identified a unique mechanism by which HIV-1 exploits C-type lectin receptors for immune evasion.

Selected Publications: 1. Grim JC, et al. “A reversible and repeatable thiol-ene bioconjugation for dynamic presentation of signaling proteins in hydrogels.” ACS Central Science, 2018. 2. Grim JC, et al. “Thiol-ene and photo-cleavage chemistry for controlled presentation of biomolecules in hydrogels.” Journal of Controlled Release, 2015. 3. Grim JC, et al. “Glycomimetic building blocks: a divergent synthesis of epimers of shikimic acid.” Organic Letters, 2011. 4. Killaars AR, Grim JC, et al. “Extended exposure to stiff microenvironments leads to persistent chromatin remodeling in human mesenchymal stem cells.” Advanced Science, 2019. 5. Aguado BA, Schuetz KB, Grim JC, et al. “Transcatheter aortic valve replacements alter circulating serum factors that mediate myofibroblast activation of valvular interstitial cells.” Science Translational Medicine, Accepted, in press, 2019.

Awards/Honors: Postdoctoral Fellowship in Cardiology, National Institutes of Health T32 (2018 – 2020) Future Faculty Scholar, American Chemical Society (2018) Postdoctoral Fellowship, Howard Hughes Medical Institute (2014 – 2017) Predoctoral Fellowship, Hirschmann Rich Award in Bioorganic Chemistry (2013) JOSHUA MICAH GROLMAN, PhD Wyss Institute, Harvard University, 58 Oxford Street, Room 200, Cambridge, MA, 02138 . [email protected]

Research Overview: Mammalian cell morphology is a key determinant of function, igniting significant interest in understanding how cell spreading is regulated by microenvironmental cues. These cues include cell-cell interactions, soluble signaling molecules, and adhesion to the extracellular matrix (ECM) via transmembrane receptors; the physical properties of the ECM, particularly its stiffness and rate of stress relaxation are known to regulate the ability of a variety of cell types to spread in 2D and 3D culture. We explore the role of matrix plasticity in controlling the spread of mesenchymal stem cells (MSCs). First, we describe a hydrogel system in which plasticity can be controlled independently of elastic modulus and stress relaxation behavior, and then demonstrate that MSC spreading and focal adhesion formation are greatest at an intermediate level of substrate plasticity. Kinetic Monte Carlo (KMC) simulations confirm cell spreading rate to be a function of the plasticity of the matrix. Matrix plasticity also affects Yes-associated protein (YAP) translocation, which can be tuned by altering actomyosin contractility. Altogether, these findings demonstrate a key role for matrix plasticity in cell spreading, and we anticipate this will have ramifications in the design of biomaterials to control cell function and fate and in numerous biological contexts where matrix properties are altered.

Education: PhD. in Materials Science and Engineering (2016) at the University of Illinois at Urbana-Champaign B.S. in Biochemistry (2011) at the University of Massachusetts at Amherst

Research/Work Experience: Postdoctoral Research Fellow at Harvard University at the Wyss Institute (June 2016-Current) NSF Graduate Research Fellow at the University of Illinois at Urbana-Champaign (May 2011-May 2016) Commonwealth College Scholar Research Fellow at the University of Massachusetts at Amherst (June 2006-Jan 2011)

Selected Publications: • Grolman, J.M.; Mansher S.; Mooney, D.J.; Eriksson, E.; Nuutila, K. “Antibiotic-containing agarose hydrogel for wound and burn care,” J. Burn Care and Research, 2019. • Koshy, S.T.; Zhang, D.K.; Grolman, J.M.; Stafford, A.G.; Mooney D.J. “Injectable nanocomposite cryogels for versatile protein drug delivery,” Acta Biomaterialia, 2018, 65, 36-43. • Montoto, E.; Gavvalapalli, N.; Hui, J.; Burgess, M.; Sekerak, N.; Hernandez-Burgos, K.; Wei, T.-S.; Kneer, M.; Grolman, J.; Cheng, K.; Lewis, J.; Moore, J.; Rodriguez-Lopez, J. “Redox Active Colloids as Discrete Energy Storage Carriers,” J. Am. Chem. Soc. 2016, 138, 13230–13237. • Grolman, J.M.; Zhang, D.; Smith, A.M.; Moore, J.S.; Kilian, K.A. "Rapid 3D Extrusion of Synthetic Tumor Microenvironments," Adv. Mater., 2015, 27, 5512-5517 (selected front cover art). • Grolman, J.M.; Inci, B.; Moore, J.S. “pH-Dependent Switchable Permeability from Core-Shell Microcapsules,” ACS Macro. Lett., 2015, 4, 441-445.

Awards/Honors: Freedom from Cancer Startup Challenge Winner 2018 UMass Boston Guest Lecturer in Biology 2017 NSF National Innovation CORPS Fellowship: Team Leader 2016 COZAD New Venture Competition Winner 2016 Fifty for The Future 2016 Illinois Innovation Prize Finalist 2016 CASSS Travel Award 2015 Illinois Conference Travel Award 2015 Intel-Racheff Award Finalist 2015, 2016 NSF IGERT Graduate Research Fellowship 2011-2013 NSF EAPSI Fellowship Awardee 2013 Commonwealth College Honors College Scholar and Research Fellow 2007-2011 Junior Fellow in the Life Sciences at UMass Amherst 2010 UMass Writing Program Outstanding Essay Award 2007 MATTHEW S. HALL, PhD Biomedical Engineering, University of Michigan, 1600 Huron Parkway, Building 520 North Campus Research Complex, Ann Arbor, MI 48105, Ann Arbor, MI, 48104 [email protected]

Research Overview: My research focuses on development of next generation cellular immunotherapies against cancer with particular focus on adoptive natural killer (NK) cell therapy. I do this by analysis of single NK cell killing dynamics in vitro and design of integrated biomaterial/CAR NK cell systems in vivo for improving therapeutic response. In project #1, I apply high content live cell imaging along with single cell omics to discover transcriptional regulators of single NK cell killing dynamics (oral presentation at BMES 2019). I show that dynamic changes in transcription factor activity in individual NK cells is associated with serial killing of cancer cells. In project #2, I develop a biomaterial implant for local delivery and activation of adoptive NK cell immunotherapy against solid tumors (poster at BMES 2019). Our in vivo results show that local delivery of NK cells on the implant improves persistence of NK cells at the site of the tumor after adoptive transfer. In project #3, I seek to understand how single cells alter their behavior in response to disease-mediated complex changes to their ECM microenvironment (Hall et al. PNAS 2016). By conducting 3D traction force microscopy across ECMs with controlled stiffness and microstructure, I show that a positive mechanical feedback loop between single Cancer cells and fibrous ECMs results in extensive stiffening, providing a new single cell mechanism for the rapid tissue stiffening associated with tumor initiation.

Education: 2016-Present: Postdoctoral Fellow, Department of Biomedical Engineering, University of Michigan, Laboratory of Lonnie Shea. 2010-2016: PhD in Biological Engineering, Cornell University, Laboratory of Mingming Wu. 2006-2010: BS Biological Engineering, Cornell University

Research/Work Experience: 2016-present: Postdoctoral Fellow, Department of Biomedical Engineering, University of Michigan, Laboratory of Lonnie Shea. 2010-2016: Graduate research assistant. Department of Biological and Environmentwl Engineering. Cornell University, Laboratory of Mingming Wu.

Selected Publications: Hall et al. Fibrous nonlinear elasticity enables positive mechanical feedback between cells and ECMs. PNAS. 2016. Hall et al. Toward single cell traction microscopy within 3D collagen matrices. Exp Cell Res. 2013. Hall and Long et al. Mapping three-dimensional stress and strain fields within a soft hydrogel using a fluorescence microscope. Biophys J. 2012. Decker, Hall et al. Design of large-scale reporter construct arrays for dynamic, live cell systems biology. ACS Syn Bio. 2018. Hall et al. Dynamic transcriptional programs controlling single NK cell killing dynamics. Cancer Research Supplement. 2019.

Awards/Honors: April 2018-Present: NIH National Research Service Award Individual Postdoctoral Fellowship (F32) 2016: Research featured in Cornell Chronicle University Press 2016: Research featured in University of Pennsylvania PennNews University Press 2016: Outstanding Teaching Assistant Cornell College of Agriculture and Life Science WOOJIN M. HAN, PhD Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30322 [email protected]

Research Overview: Musculoskeletal disorders and injuries are the leading contributors to disability worldwide. To this end, my research goal is to establish translational technologies for musculoskeletal soft tissue regeneration by controlling cell-matrix interactions. My doctoral work established how mechanical strain exerted at the tissue-level is progressively transferred to the underlying cells in musculoskeletal soft tissues. In doing so, I revealed that the cells within the native tissues are subjected to “strain-shielded” environment, where the levels of strain attenuation and stretch-induced cellular responses are dependent on tissue type, local structure, composition, age, and pathology. Additionally, in close collaboration with Dr. Robert Mauck, we developed a tunable tissueengineered platform that mimics the structure, mechanical and cellular functions of aging fibrocartilage across multiple length scales. As a postdoctoral fellow, I engineered a synthetic designer matrix for facilitating muscle stem cell transplantation in skeletal muscles. The engineered matrix supports muscle stem cell function and significantly enhances cellular engraftment efficiency upon transplantation. Moving forward, my research program will synergize concepts of biomaterials, tissue engineering, biomechanics, and cell biology to systematically investigate how cells interact with their surrounding niche in regulating musculoskeletal soft tissue homeostasis and pathogenesis. These findings will be further applied to engineer new regenerative technologies for treating injuries and diseases pertaining to the musculoskeletal system by harnessing tissue-specific cell-matrix interactions.

Education: • Ph.D., Bioengineering, University of Pennsylvania (2015) • M.S.E., Bioengineering University of Pennsylvania (2010) • B.S., Biomedical Engineering, University of Rochester (2009)

Research/Work Experience: • Postdoctoral Fellow, Georgia Tech | Advisors: Andrés J. García, Ph.D. & Young C. Jang, Ph.D. (2015 – Present) • Graduate Student/Researcher, University of Pennsylvania | Advisor: Dawn M. Elliott, Ph.D.(2009 – 2015) • Undergraduate Researcher, University of Rochester | Advisor: Michael R. King, Ph.D. (2008 – 2009)

Selected Publications: • Han, WM, et al. “Co-delivery of Wnt7a and Muscle Stem Cells using Synthetic Bioadhesive Hydrogel Enhances Murine Muscle Regeneration and Cell Migration during Engraftment” Acta Biomaterialia, (2019), 96, 243-252. • Han, WM, et al. “Synthetic Matrix Enhances Transplanted Satellite Cell Engraftment in Dystrophic and Aged Skeletal Muscle with Comorbid Trauma” Science Advances, (2018), 4:eaar4008. • Han, WM‡; Heo, SJ‡, et al. “Microstructural Heterogeneity Directs Micromechanics and Mechanobiology in Native and Engineered Fibrocartilage” Nature Materials, (2016), 15(4), 477-484. ‡Equal contribution. • Han, WM, et al. “Macro- to Microscale Strain Transfer in Fibrous Tissues is Heterogeneous and Tissue Specific” Biophysical Journal, (2013), 105(3), 807-817. • Han, W‡; Allio, BA‡, et al. “Nanoparticle Coatings for Enhanced Capture of Flowing Cells in Microtubes” ACS Nano, (2010), 4(1), 174-180. ‡Equal contribution.

Awards/Honors: • NIH/NHLBI Ruth L. Kirschstein F32 Postdoctoral Fellowship (2018) • Communications Chair Elect, Society for Biomaterials – Young Scientist Group (2020 – 2021) • Co-Chair, Gordon Research Seminar, STEEM (2018 – 2020) • Outstanding Oral Presentation Award, 2nd Annual Georgia CTSA Conference (2019) • Outstanding Poster Award, Gordon Research Conference, STEEM (2018) • PMSE Future Faculty Scholar, The American Chemical Society, PMSE Division (2018) • 1st Place Poster Presentation Award, Regenerative Medicine Workshop (2018) • Glenn/AFAR Postdoctoral Fellowship for Translational Research on Aging (2015) • Student/Fellow Award, BMES Cellular and Molecular Bioengineering Conference (2015) • Best Poster Presentation Award, Philadelphia Spine Research Symposium (2012) • Best Podium Presentation Award, 9th CBER Biomechanics Research Symposium, Univ of Delaware (2012) • Charles L. Newton Prize, University of Rochester (2009) MAHDI HASANI, PhD Bioengineering, UCLA, 410 Westwood Plaza, UCLA Eng V, Los Angeles, CA, 90095 - [email protected]

Research Overview: • Drug Delivery and Nanoparticles: Development of polymeric nanoparticles as cancer and stem cell regulators. • Biomaterials and Hydrogels: Development of hydrogel biomaterials to engineer stem cell fate. • Microfluidics and Nanomanufacturing: Regulation of physiochemical properties of polymeric nanoparticles via engineered microfluidic platforms. • Fuel Cell and NanoEnergy: Design of proton exchange membranes for hydrogen and direct methanol fuel cell applications.

Education: • Georgia Institute of Technology, Dec 2017. Ph.D. Bioengineering • Amirkabir University of Technology, June 2013. Ph.D. • École Polytechnique Fédérale de Lausanne (EPFL), June 2012. M.Eng. Bioengineering • Amirkabir University of Technology, June 2009. M.Sc. Polymer Engineering • Amirkabir University of Technology, Dec 2009. B.Sc. Biomedical Engineering • Amirkabir University of Technology, Dec 2007. B.Sc. Polymer Engineering

Research/Work Experience: • UCLA Bioengineering, 2018-Present; Assistant Project Scientist – Immunoengineering. Advisors: Song Li, PhD; Manish Butte, MD PhD; Paul S. Weiss, PhD • UCLA School of Dentistry, 2018-Present; Assistant Project Scientist – Dental Biomaterials. Advisors: Alireza Moshaverinia, DDS PhD; Tara Aghaloo, MD DDS PhD • Georgia Tech Bioengineering, 2014-2017 Graduate Research Assistant. Advisors: Karl I. Jacob, PhD; Mostafa El-Sayed, PhD • Amirkabir University of Technology, Biomedical Engineering, 2013-2014; Assistant Professor • Royan Institute, Cell Engineering; 2013-2014; Courtesy Faculty Member • EPFL Center of MicroNanoTechnology (CMi), 2012-2013; Senior Engineer. Advisor: Phillipe Renaud, PhD • EPFL Institute of Bioengineering, 2011-2012; Graduate Researcher. Advisor: Jeffrey A. Hubbell, PhD • Swiss Institute for Experimental Cancer Research (ISREC), 2011-2012; Graduate Researcher. Advisor: Douglas Hanahan, PhD • Hydrogen & Fuel Cell Inc., 2007-2010, R&D member: Renewable Energy Educational Kits. CEO: Homayoun Moaddel, PhD

Selected Publications: 1. Hasani, et al. Hierarchically Patterned Polydopamine-Containing Membranes for Periodontal Tissue Engineering. ACS Nano. 13(2019)3830. 2. Hasani, et al. Mechanobiological Mimicry of Helper T Lymphocytes to Evaluate Cell-Biomaterial Cross-Talk. Advanced Materials. 30(2018)1706780. 3. Majedi, Hasani, et al. Cytokine secreting microparticles engineer the fate and the effector functions of T cells. Advanced Materials. 30(2018)1703178. 4. Hasani, et al. Microfluidic Manipulation of Core/Shell Nanoparticles for Oral Delivery of Chemotherapeutics: A New Treatment Approach for Colorectal Cancer. Advanced Materials. 28(2016)4134. 5. Hasani-Sadrabadi, et al. Microfluidic-Assisted Self-Assembly of Complex Dendritic Polyethylene Drug Delivery Nanocapsules. Advanced Materials. 26(2014)3118.

Awards/Honors: • Early Career Board of ACS Biomaterials Science & Engineering, ACS. 2018. • Sigma Xi Best Ph.D. Thesis Award, Georgia Tech. 2018. • Image of the Year Award, Southern California Society for Microscopy and Microanalysis. 2017. • Wang Outstanding Young Nanoscience Researchers Award. 2015. • J.W. Richards Summer Fellowship Award, ECS. 2015. • ACS CELL Division Graduate Student Award, ACS. 2015. • Electrochemical Society Student Achievement Award, ECS. 2015. • Young Khwarizmi Award, 3rd (Fundamental Research, 2013), 2nd (Applied Research, 2012), 3rd (Innovation, 2005). • Grand Prize Winner, Baker Award for Fuel Cell Research. 2011. • 2nd Winner, Swiss Micronanotechnology competition. 2011. • Excellence Scholarship. EPFL. 2010-11. • 1st Winner, Distinguished Polymer Science Alumni Award, IPS. 2010. • Eni Award Finalist. 2010. • Student of the Year in Engineering Presidential Award. 2008. • National Scientific Prize, INEF. 2007. • Gold Medal in Invention, SIIF. 2006. • Invention Diploma. IENA. 2006. HODA HATOUM, PhD Postdoctoral fellow, Biomedical Engineering Department, The Ohio State University, Columbus, Ohio, 43210 [email protected]

Research Overview: During my PhD, I was involved in cutting-edge research advancing the knowledge of surgical and transcatheter heart valves using high-fidelity experimental techniques such as particle image velocimetry and patient specific 3D modeling among others. The major findings of my work were published in over 12 high impact journal articles. The patient specific models I developed even attracted national press coverage. During my postdoc, I differentiated beyond my doctoral research by implementing design modifications on the surface of mechanical valve leaflets in order to control the flow. Superhydrophobic materials are an ongoing project that falls under mechanical valve flow control. In order to improve hemodynamics, durability and valve performance, implementing novel materials is becoming necessary. New materials are on the rise specifically in the clinical realm as the search for the optimal material with improved interface properties is ongoing. Thus, the combination of vortex generators (passive flow control) along with superhydrophobic material will improve fluid-solid interface and flow behavior. This study has resulted in several publications and submitted manuscripts in addition to the American Heart Association postdoctoral Fellowship award. In addition, my work includes the implementation of novel transcatheter manipulation techniques in order to improve hemodynamics and minimize adverse effects (BASILICA, BISILICA). Anomalous coronary arteries in infants and children in collaboration with Nationwide Children’s hospital is also an ongoing long-term project. The collaboration with Nationwide children’s hospital has given me new ideas regarding immediate projects particularly in the areas of Fontan and MBT-shunt procedures. My postdoctoral work resulted in more than 9 publications so far.

Education: •The Ohio State University, August 2018. PhD, Mechanical Engineering • American University of Beirut, June 2009. BE, Mechanical Engineering

Research/Work Experience: • The Ohio State University, Biomedical Engineering Department, 2018 - present Postdoctoral Fellow; Advisor: Lakshmi Prasad Dasi, PhD • The Ohio State University, 2015 – 2018 Graduate Research Assistant, Mechanical Engineering; Advisor: Lakshmi Prasad Dasi, PhD • Dar Group, 2009 – 2014 Mechanical Design Engineer (consultant)

Selected Publications: 1. Hatoum, Hoda, et al. "Impact of leaflet laceration on transcatheter aortic valve-in-valve washout: BASILICA to solve neosinus and sinus stasis." JACC: Cardiovascular Interventions 12.13 (2019): 1229-1237. 2. Hatoum, Hoda, et al. "An in vitro evaluation of turbulence after transcatheter aortic valve implantation." The Journal of thoracic and cardiovascular surgery 156.5 (2018): 1837-1848. 3. Hatoum, Hoda, and Lakshmi P. Dasi. "Reduction of pressure gradient and turbulence using vortex generators in prosthetic heart valves." Annals of biomedical engineering 47.1 (2019): 85-96. 4. Hatoum, Hoda, et al. "Effect of severe bioprosthetic valve tissue ingrowth and inflow calcification on valve-in-valve performance." Journal of biomechanics 74 (2018): 171-179. 5. Hatoum, Hoda, et al. "Stented valve dynamic behavior induced by polyester fiber leaflet material in transcatheter aortic valve devices." Journal of the mechanical behavior of biomedical materials 86 (2018): 232-239. 6. Hatoum, Hoda, et al. "Implantation depth and rotational orientation effect on valve-in-valve hemodynamics and sinus flow." The Annals of thoracic surgery 106.1 (2018): 70-78.

Awards/Honors: • Biomedical Engineering Society Career Development Award (2019). • ASME-BED SPC: BS Level Awards (2nd place) “A Study of Pressure Dynamics across a Stenotic Orifice” by Tori Burton, Hoda Hatoum and Lakshmi Prasad Dasi (2019). • Presidential Fellowship in the Ohio State University (2018). • Runner up in the ASME-BED PhD Level Student Paper Competition (2018). • World Congress of Biomechanics Diversity travel award recipient (2018). • Mimics Innovation Award first prize winner in America region (2018). CARL D. HERICKHOFF, PhD Radiology, Stanford University, 3155 Porter Dr, Palo Alto, CA, 94304 . [email protected]

Research Overview: My biomedical ultrasound research spans both ‘fundamental’ and ‘applied’ ultrasound engineering topics: creating novel devices and methods to obtain unique signals and produce new imaging information, and developing new approaches to integrate and translate ultrasound into use in challenging healthcare settings. My lab will have two broad areas of focus: (1) specialized ultrasound imaging systems, and (2) novel ultrasound transducer technologies. Current and future projects in these areas include: -Large-scale ultrasound body scanner array and associated imaging algorithms -Augmented ultrasound acquisition and visualization for enhanced clinical use -Cylindrical transducer arrays for endolumenal elasticity imaging and therapy -Dual-frequency transducer arrays for superharmonic contrast imaging -Paneled-cone array for quantitative lactation measurement

Education: Ph.D. Biomedical Engineering, 2011, Duke University M.S. Biomedical Engineering, 2009, Duke University B.S. Electrical Engineering, 2005, University of Notre Dame

Research/Work Experience: 2018-pres. - Deputy Director, Radiological Sciences Laboratory, Stanford University School of Medicine, CA 2015-pres. - Research Engineer, Radiology, Stanford University School of Medicine, CA 2013-2015 - Research Scientist and Instructor, Biomedical Engineering, Duke University, NC 2011-2013 - Advanced Transducer R&D Engineer, Philips Healthcare, Andover, MA 2011 - Postdoctoral Research Assistant, Dept. of Biomedical Engineering, Duke University, NC 2010 - Ultrasound R&D Graduate Intern, General Electric Global Research, Niskayuna, NY

Selected Publications: Herickhoff CD, Grant GA, Britz GW, and Smith SW. Dual-mode IVUS catheter for intracranial image-guided hyperthermia: feasibility study. IEEE Transactions on Ultrasonics, Ferroelectrics, and Frequency Control, 57.11 (2010): 2572-2584. doi:10.1109/ULTSYM.2010.5935594 Herickhoff CD, Wilson CM, Grant GA, Britz GW, Light ED, Palmeri ML, Wolf PD, and Smith SW. Dual-mode IVUS transducer for image-guided brain therapy: preliminary experiments. Ultrasound in Medicine and Biology, 37.10 (2011): 1667-1676. doi: 10.1016/j.ultrasmedbio.2011.06.017 Broder JS, Jaffa EJ, Morgan MR, Herickhoff CD, Smith BP, Peethumnongsin E, Dahl JJ. Brain Imaging Using a Novel Three- Dimensional Ultrasound System. Annals of Emergency Medicine, 70.4 (2017): S154-S15. doi: 10.1016/j.annemergmed.2017.07.364 Herickhoff CD, Morgan MR, Broder JS, and Dahl JJ. Low-cost Volumetric Ultrasound by Augmentation of 2D Systems: Design and Prototype. Ultrasonic Imaging, 40.1 (2018): 35-48. doi: 10.1177/0161734617718528 Morgan MR, Broder JS, Dahl JJ, and Herickhoff CD. Versatile Low-cost Volumetric 3D Ultrasound Platform for Existing Clinical 2D Systems. IEEE Transactions on Medical Imaging, 37.10 (2018): 2248-2256. doi: 10.1109/TMI.2018.2821901 Broder JS, Jaffa E, Irons T, Peethumnongsin E, Theophanous R, Limkakeng AT, Vissoci JR, Morgan MR, Herickhoff CD, Smith BP, Dahl JJ. 3D Volume Ultrasound of the Right Lower Quadrant for Diagnosis of Appendicitis. Annals of Emergency Medicine, 72.4 (2018): S85. doi: 10.1016/j.annemergmed.2018.08.217 Di Ianni T, Bose RJC, Sukumar UK, Bachawal S, Huaijun Wang, Telichko A, Herickhoff CD, Robinson E, Baker S, Vilches-Moure JG, Felt SA, Gambhir SS, Paulmurugan R, Dahl JJ. Ultrasound and microbubble-mediated targeted delivery of therapeutic microRNA-loaded nanocarriers to deep liver and kidney tissues in pigs. Journal of Controlled Release (2019). doi: 10.1016/j.jconrel.2019.07.024 C. D. Herickhoff and J. J. Dahl. Advances in Medical Physics – 2016, volume 6, chapter 4: Advances in Ultrasonic Imaging Technology, pages 71–96. Medical Physics Publishing, Madison, WI, 2016.

Awards/Honors: Student Paper Competition Finalist - IEEE International Ultrasonics Symposium, 2010. ANDREW W. HOLLE, PhD Cellular Biophysics Dept., Max Planck Institute for , Heisenbergstrasse 3, 70569 Stuttgart, Germany [email protected]

Research Overview: My research at the Max Planck Institute for Medical Research and the University of California, San Diego has primarily focused on utilizing bioengineering strategies to identify and exploit the mechanosensitive mechanisms underpinning cell behavior, including stem cell differentiation, cancer cell invasion, and cytoskeletal adaptation. During my doctoral research, I identified the role that the focal adhesion protein vinculin plays in mechanosensitive stem cell myogenesis. Expanding on this principle, I identified a number of novel focal adhesion-based mechanotransduction pathways in stem cells using high content image analysis techniques. For my postdoctoral research, I demonstrated that cancer cells are capable of dynamically switching from mesenchymal to amoeboid migration in response to self-imposed confinement, and that this process is independent of ECM composition. Using a custom-designed microfluidic chip, I measured dynamic changes in the cytoskeleton during extreme confinement, and determined that the two-stage transition is initially Rac1-dependent. In addition, I led an international team in the development of a continuous stiffness gradient hydrogel platform compatible with high content image analysis. This platform provides the highest resolution look into substrate stiffness-induced mechanobiology ever attained, capable of monitoring mechanosensitive cell behavior in over 10,000 cells per well and subsequently mapping the resultant output onto a continuous axis of substrate stiffness. My long term research goal is to leverage innovative microfluidic biochips, nanoscale biomaterials, and to understand cell behavior in response to physiologicallyrelevant, multidimensional physical confinement. Applications for this work range from basic science aimed at elucidating the complex mechanisms underlying mechanotransduction to translational clinical approaches for precision medicine to the development of commercially viable tissue engineering products.

Education: • University of California, San Diego, December 2013, Ph.D., Bioengineering • Arizona State University, June 2008, B.S.E., Biomedical Engineering

Research Experience: • Max Planck Institute for Medical Research, 2017 – present Max Planck Institute Postdoctoral Fellow, Dept. of Cellular Biophysics; Advisor: Joachim P. Spatz, Ph.D. • Max Planck Institute for Intelligent Systems, 2014-2016 Max Planck Institute Postdoctoral Fellow, Dept. of New Materials and Biosystems; Advisors: Joachim P. Spatz, Ph.D. and Ralf Kemkemer, Ph.D. • University of California, San Diego, 2008-2013 Graduate Research Assistant Bioengineering; Advisor: Adam J. Engler, Ph.D. • Biodesign Institute, 2007-2008 Undergraduate Research Assistant Center for Ecogenomics; Advisor: Deirdre Meldrum, Ph.D. • Arizona State University, 2005-2008 Undergraduate Research Assistant Biomedical Engineering; Advisor: Christine Pauken, Ph.D.

Selected Publications: 1. Holle, A.W., Govindankutty Devi, N., Clar, K., Fan, A., Kemkemer, R., Spatz, J.P. “Cancer cells invade confined microchannels via a self-directed mesenchymal-to-amoeboid transition” Nano Letters 19, 2280-2290 (2019) 2. Hadden, W.*, Young, J.L.*, Holle, A.W.*, McFetridge, M., Kim, D.Y., Wijesinghe, P., Taylor-Weiner, H., Wen, J.H., Lee, A.R., Bieback, K., Vo, B.N., Sampson, D.D., Kennedy, B.F., Spatz, J.P., Engler, A.J., Choi, Y.S. “Stem cell migration and mechanotransduction on linear stiffness gradient hydrogels” PNAS 114, 5647-5652 (2017) *Equal contribution 3. Holle, A.W., Kalafat, M., Sales Ramos, A., Seufferlein, T., Kemkemer, R., Spatz, J.P. “Intermediate filament reorganization dynamically influences cancer cell alignment and migration” Scientific Reports 7, 45152 (2017) 4. Holle, A.W., McIntyre, A.J., Kehe, J., Wijesekara, P., Young, J.L., Vincent, L.V., Engler, A.J. “High content image analysis of focal adhesion-dependent mechanosensitive stem cell differentiation” Int. Biology 8, 1049-1058 (2016) 5. Holle, A.W., Tang, X., Vijayraghavan, D., Vincent, L.G., Fuhrmann, A., Choi, Y.S., del Alamo, J.C., and Engler, A.J. “In situ mechanotransduction via vinculin regulates stem cell differentiation” Stem Cells 31, 2467-2477 (2013)

Awards/Honors: American Association for Cancer Research (AACR) Basic Research Fellow, 2016-2017 Max Planck Institute Postdoctoral Fellowship, 2014-2017 Chancellor’s Dissertation Medal, Department of Bioengineering Nominee University of California, San Diego, 2013 Shu Chien NSF-BMES Award for Excellence in Mechanobiology and Mechanotransduction, 2011 QUANYIN HU, PhD Koch Institute for Integrative Cancer Research, MIT, 500 main street, building 76-632B, Cambridge, MA, 02139 [email protected]

Research Overview: My research focus is at the interface of biomaterials and physiology for drug delivery and immunotherapy. I am particularly interested in exploring novel strategies that mimic natural cells and leverage physiological conditions for delivering therapeutics in spatial- and temporal-controlled manners. In my previous studies, by harnessing the interaction between platelet and cancer cells, I developed a platelet-mimicking platform to enhance the delivery efficacy of various therapeutics against multiple diseases including breast cancer, multiple myeloma, and thrombus. Later on, I further explored the application of platelet-based platform by integrating with immune checkpoint inhibitor antibodies to increase the objective response rate and treatment efficacy of checkpoint blockade/CAR-T cellsbased cancer immunotherapy and mitigate the side effects. Currently, I am using the macro/nanofabrication technology to design the drug delivery device with distinctive pulsatile-release kinetics to tackle the effective therapeutic window between different treatment regimens against cancer.

Education: Ph.D., Biomedical Engineering, University of North Caroline at Chapel Hill and North Carolina State University, 2014-2018 M.S., Pharmaceutics, Fudan University, 2011-2014 B.S., Pharmaceutical Engineering, China Pharmaceutical University, 2007-2011

Research/Work Experience: 2018-present, Postdoc Associate, Advisor: Dr. Robert S. Langer, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology 2014-2018, Graduate research assistant, Advisor: Dr. Zhen Gu, Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University

Selected Publications: 1. Quanyin Hu, Qian Chen, Huitong Ruan, Sarah Ahn, Elena Dukhovlinova, Edikan Archibong, Kang Yang, Di Wen, Gianpietro Dotti* and Zhen Gu*. “Cell Delivery Reservoir Promotes Immune Cell Antitumor Activity.” Nature Biomedical Engineering, In revision, 2019. 2. Quanyin Hu, Wujin Sun, Jinqiang Wang, Huitong Ruan, Xudong Zhang, Yanqi Ye, Song Shen, Chao Wang, Weiyue Lu, Ke Cheng, Gianpietro Dotti, Joshua F. Zeidner, Jun Wang, Zhen Gu*. "Conjugation of Haematopoietic Stem Cells and Platelets Decorated with Anti-PD-1 Antibodies Augments Anti-Leukaemia Efficacy." Nature Biomedical Engineering, 2, 2018. 3. Quanyin Hu, Wujin Sun, Chenggen Qian, Hunter Bomba, Hongliang Xin , Zhen Gu*. “Relay Drug Delivery for Amplifying Targeting Signal and Enhancing Anticancer Efficacy”. Advanced Materials, 29, 2017. 4. Quanyin Hu, Chenggen Qian, Wujin Sun, Jinqiang Wang, Zhaowei Chen, Hunter Bomba, Hongliang Xin, Qundong Shen, Zhen Gu*. “Engineered Nanoplatelets for Enhanced Treatment of Multiple Myeloma and Thrombus”. Advanced Materials, 28, 2016. 5. Quanyin Hu, Wujin Sun, Yue Lu, Hunter Bomba, Yanqi Ye, Tianyue Jiang, Ari Isaacson, Zhen Gu*. “Tumor MicroenvironmentMediated Construction and Deconstruction of Extracellular Drug-Delivery Depots”. Nano Letters, 16, 2016. 6. Quanyin Hu, Wujin Sun, Chenggen Qian, Chao Wang, Hunter Bomba, Zhen Gu*. “Anticancer Platelet-Mimicking Nanovehicles”. Advanced Materials, 27, 2015. 7. Huitong Ruan#, Quanyin Hu#, Di Wen, Qian Chen, Guojun Chen, Yifei Lu, Jinqiang Wang, Hao Cheng, Weiyue Lu*, Zhen Gu*. “A Dual-Bioresponsive Drug Delivery Depot for Immune Checkpoint Blockade”. Advanced Materials, 31, 2019. (# equal contribution).

Awards/Honors: BMES Career Development Award, 2019 AAPS Best Abstract Award, 2019 Vasculata Scholarship, 2019 Chinese Government Award for Outstanding Ph.D. Students Abroad, Washington, DC, 2018 The Excellent Master Dissertation of Shanghai Municipality, Shanghai, 2015 Graduate with honor of Shanghai (first-class honor), Shanghai, 2014 XIAO HU, PhD Biomedical Engineering, University of Virginia, 415 Lane Road, MR5 Room 2231, Charlottesville, VA, 22903 . [email protected] Research Overview: Millions of Americans suffer from neuromusculoskeletal disorders, such as muscular dystrophy, cerebral palsy, stroke, etc. Undoubtedly, human neuromusculoskeletal system is at the center for understanding, diagnosis, treatment planning and prognosis of neuromusculoskeletal disorders. However, limitations of experimental approach alone make it difficult to quantitatively assess how the neuromusculoskeletal system responds to disorders and the related treatments. When personalized with patient-specific data, multiscale neuromusculoskeletal simulation with its non-invasive nature will provide a critical tool for elucidating mechanisms of disorders and guiding treatment planning. My overall research goal is to apply multiscale, personalized neuromusculoskeletal modeling and simulation to understand the mechanisms of neuromusculoskeletal disorders and develop rehabilitative and therapeutic strategies for limiting their impact on patients and society. In my PhD work, using combined neuromusculoskeletal simulation and experimental approaches, I revealed the neural and biomechanical mechanisms that the central nervous system uses to regulate limb posture and force, two essential requirements for completing any functional tasks, in the presence of uinexpected perturbations. These findings are important for understanding how injury and disease to the neuromusculoskeletal system alter the ability to regulate arm mechanics in a functionally appropriate manner. To develop my experience towards clinically orientated, collaborative research, my postdoctoral work applied multiscale neuromusculoskeletal simulations to examine the pathophysiological mechanisms that lead to impairments and functional deficits in devastating neuromusculoskeletal disorders, such as Duchenne muscular dystrophy (DMD) and volumetric muscle loss injury. Currently, supported by the NIH grant R21AR068562, I am integrating imaging, strength and gait measurements to create patientspecific dynamic simulations of walking that examine the relationship between muscle function in locomotion and muscle degeneration in DMD. As a junior faculty, I will leverage my experiences to build a multi-disciplinary research program that will: 1) develop a framework for fast development of multiscale, personalized neuromusculoskeletal models; and 2) utilize simulations to guide the development of rehabilitative strategies that prolong walking in patients with DMD.

Education: • PhD, Biomedical Engineering, Dec 2012, Northwestern University, Evanston, IL • MS, Mechanical Engineering, May 2007, Tsinghua University, Beijing, China • BS, Measurement and Control Technology and Instruments, May 2004, Tsinghua University, Beijing, China

Research/Work Experience: • Postdoctoral Research Associate, 2013 – Present, University of Virginia, BME, Advisor: Silvia Blemker, PhD • Postdoctoral Research Fellow, 2012 – 2013, Rehabilitation Institute of Chicago, SMPP, Advisor: Wendy Murray, PhD • Graduate Research Assistant, 2007 – 2012, Northwestern University, BME, Advisors: Eric Perreault, PhD & Wendy Murray, PhD

Selected Publications: 1. Passipieri JA*, Hu X*, Mintz E, Dienes J, Baker HB, Wallace CH, Blemker SS, and Christ GJ (2019). In Silico and In Vivo Studies Detect Functional Repair Mechanisms in a Volumetric Muscle Loss Injury. Tissue engineering Part A. (*contributed equally) 2. Hu X, Charles JP, Akay T, Hutchinson JR and Blemker SS (2017). Are Mice Good Models For Human Neuromuscular Disease? Comparing Muscle Excursions in Walking Between Mice and Humans. Skeletal Muscle 7: 26. 3. Hu X, and Blemker SS (2015). Musculoskeletal simulation can help explain selective muscle degeneration in Duchenne muscular dystrophy. Muscle and Nerve 52(2), 174-182. 4. Hu X, Murray WM and Perreault EJ (2012). Biomechanical constraints on the feedforward regulation of endpoint stiffness. Journal of Neurophysiology 108(8), 2083-2091. 5. Hu X, Murray WM and Perreault EJ (2011). Muscle short-range stiffness can be used to estimate the endpoint stiffness of the human arm. Journal of Neurophysiology 105(4), 1633-1641.

Awards/Honors: • NIH/NIAMS R21 Grant (R21AR068562), 2016-2019, National Institutes of Health • SEAS Post-Doctoral Teaching Fellowship, 2018, School of Engineering & Applied Science, University of Virginia • NCSRR/OpenSim Visiting Scholarship, 2016, National Center for Simulation in Rehabilitation Research, Stanford University • ISB Student International Travel Award, 2011, International Society of Biomechanics KAI HUANG, PhD Biomedical Engineering, Northwestern University, 2170 Campus Drive, Evanston, IL, 60208 . [email protected]

Research Overview: My research sits at the intersection of biomaterials, biophysics and systems biology, with the overarching goal of using theoretical and computational tools to guide biomedical design and genomic engineering. During my doctoral study, I conducted molecular dynamics simulations to show how ionic context shapes the hydrophobic interaction that drives the self-assembly of biomolecules and artificial amphiphiles. I also developed a non-equilibrium statistical theory of flow boundary condition, which is critical to understand the fluidic behavior in nanofluidic channels. The method provides a powerful numerical tool to evaluate liquid-solid friction and slip through computer simulations. My postdoctoral research has focused on revealing the sequence-structure-function relationship of intrinsically disordered proteins inside the nuclear pore. To tackle this largest molecular channel inside eukaryotic cells, I have built a computational microscope capable of visualizing the functional core of the nuclear pore complex that has remained a long-standing challenge for experimental characterization. Inspired by the nuclear pore, I have also designed artificial nanopores based on stimuliresponsive amphiphilic copolymers. My current research focuses on the self-organization of 3D genome, an emerging challenge in biology that has attracted intensive research energy in the last decade. Working closely with experimental collaborators, I have developed a new model of chromatin folding, which explains a wide array of experimental observations from genomic contact scaling to heterogeneous chromatin packing. My future research program will further explore the nuclear structure, with specific interests in how the genetic and epigenetic codes direct the folding of interphase DNA. I will also integrate my multiscale modeling skills with machine learning to design bio- inspired nanomaterials and develop new strategies to treat systems biology diseases.

Education: Ph. D., 2015, Materials Science, Unversity of Wisconsin-Madison, WI, USA B. Eng., 2009, , Tsinghua University, Beijing, China

Research/Work Experience: 2015-present: Postdoctoral Fellow in the group of Prof. Igal Szleifer, Biomedical Engineering Department, Northwestern University 2009-2015: Gruaduate Research Assistant in the Computational Materials Group of UW-Madison, Advisor: Prof. Izabela Szlufarska

Selected Publications: • K. Huang*, Y. Li, A. Shim, R. Virk, V. Agrawal, A. Eshein, R. Nap, L. Almassalha, V. Backman* and I. Szleifer*, “Physical and data structure of 3D genome”, bioRxiv, doi: https://doi.org/10.1101/596262 (2019), under review of Science Advances (*co-corresponding author) • K. Huang, M. Tagliazucchi, S.H. Park, Y. Rabin and I. Szleifer, “Molecular model of the nuclear pore complex reveals a thermoreversible FG-network with distinct territories occupied by different FG motifs”, bioRxiv, doi: https://doi.org/10.1101/568865 (2019), under review of Biophysical Journal • M. Tagliazucchi, K. Huang and I. Szleifer, “Routes for nanoparticle translocation through polymer-brush-modified nanopores”, Journal of Physics: Condensed Matter, 30, 274006 (2018) • K. Huang and I. Szleifer, “Design of multifunctional nanogates in response to multiple external stimuli using amphiphilic copolymer”, Journal of the American Chemical Society, 139, 6422-6430 (2017) • K. Huang, S. Gast, C. D. Ma, N. Abbott and I. Szlufarska, “Comparison between the free and immobilized ion effects on hydrophobic interactions: a molecular dynamics study”, The Journal of Physical Chemistry B, 119, 13152-13159 (2015) • K. Huang* and I. Szlufarska*, “Effect of interfaces on nearby Brownian motion”, Nature Communications, 6:8558 doi: 10.1038/ncomms9558 (2015) (*co-corresponding author) • K. Huang* and I. Szlufarska*, “Green-Kubo relation for friction at liquid-solid interfaces”, Physical Review E 89, 032119 (2014) (*co-corresponding author) • K. Huang and I. Szlufarska, “Friction and slip at solid-liquid interface in vibrational systems”, Langmuir 28, 17302 (2012)

Awards/Honors: • GLCACS Research Presentation Third Place, 2017 • TTMS Summer School Best Poster Award, 2014 • First-class Academic Scholarship of Tsinghua University, 2008 • Second-class Academic Scholarship of Tsinghua University, 2006 HUAN-HSUAN HSU, PhD 1Tufts University, 4 Colby St, Medford, MA, 02155, 2BME, Tufts University, 4 Colby St, Medford, MA, 02155 [email protected]

Research Overview: My future research awill first be focused on integrating my expertise in sensors and bioelectronics to create an innovative platform for fabricating functional bioelectronic devices. In my research statement, I present my research initiatives centred on: (i) Microbe derived bio- electronic- interfaces: programing the growth of conductive protein based structures from EAB networks as building blocks for bioelectronic device fabrication; (ii) Bioenabled sensors: Exploiting the native and genetically engineered conductive protein as the biosensor to enable direct and effective transduction from biological signals to electrical signals; and (iii) Bioenabled cellular electronic devices: Creating the ECM- and BNW- based microelectrodes and field-effect transistors to seamlessly interface the electrical transducer and targeted biological components for biological signal recording and manipulations.

Education: 2010-2015 Ph.D. at School of Biomedical Engineering, McMaster University 2008-2010 M.A. Sc. of Chemical Engineering, McMaster University, 2005-2007 M. Sc. of Environmental Engineering, National Cheng Kung University 2001-2005 B. Sc of Environmental Engineering, National Cheng Kung University

Research/Work Experience: 2016-current Tufts University, Department of Biomedical Engineering Functional bio-derived materials for Bioelectronics, Bioenergy, Biosensing and Environmental Applications Supervisor: Prof. Xiao Cheng Jiang 2010-2016 McMaster University Micro-sensing array for water monitoring 2008-2010 McMaster University Particle Movement in Paper Porous Media: Influence Factors and Model 2005-2007 National Cheng Kung University Nanosize ZnO thin films VOCs sensor

Selected Publications: 1.H. H. Hsu, X. Jiang (2019) ““Living” Inks for 3D Bioprinting” Cell Press: Trends in Biotechnology https://doi.org/10.1016/j.tibtech.2019.04.014 2. L. Hsu, P. Deng, Y. Zhang, X. Jiang (2018) “Core/Shell Bacterial Cables: A One-Dimensional Platform for Probing Microbial Electron Transfer” Nano letters, 18 (7), pp 4606–4610 3. H. H. Hsu, P. Deng, Y. Zhang, H. N. Nguyen, X. Jiang (2018) “Nanostructured interfaces for probing and facilitating extracellular electron transfer” J. Mater. Chem. B, 6, 7144-7158 4. Amit Kumar*, L.H.H. Hsu et al. (2017) “The ins and outs of microorganism–electrode electron transfer reactions” Nature Review Chemistry, vol. 1 pp. 24 5. L.H.H. Hsu, E. Houqe, P.R. Selvaganapathy*, P. Kruse* (2015) “A Carbon Nanotube based Resettable Sensor for Measuring Free Chlorine in Drinking Water” Applied Physics Letter 106, 063102

Awards/Honors: 2016-2018 NSERC Postdoctoral Fellowship 2012-2013 Ontario Graduate Scholarship 2012 School of graduated Studies Grant in Aid for Travel and Field Work 2006-2007 Department Outstanding M.S. student 2005-2006 Arounding-You Environmental Eng. Co. Scholarship JUN ISHIHARA, PhD Pritzker school of , University of Chicago 5640 S Ellis Ave 301, 60637 IL USA (Tel: +17734498582 E–mail: [email protected])

Research Overview: I am highly multidisciplinary having trained in immunotherapy, regenerative medicine, pharmacology, toxicology, stem cell biology, and protein engineering. I have recently developed exciting protein-engineering based new drug delivery systems for targeting cancer, skin wound regeneration, autoimmune diseases (i.e. rheumatoid arthritis, diabetic pancreas, experimental autoimmune encephalomyelitis, and inflammatory bowel disease), and fibrosis. We have engineered extracellular matrix-binding protein drugs, to retain therapeutics around the disease sites, which would solve unmet clinical needs and holds translational potential. My team aims to directly address current issues in cancer immunotherapy by developing new protein therapeutics through the drug delivery systems and by acquiring biological knowledge. I aim to develop a strong research laboratory at the interface of my two key areas of expertise – tissue regeneration and immunoengineering.

Postdoctoral scholar (Jeffrey A. Hubbell, Ph.D. – Research Advisor) University of Chicago, Pritzker School of Molecular Engineering Feb, 2016-present Ecole Polytechnique Fédérale de Lausanne, Bioengineering May, 2014-Feb, 2016 Ph.D. course (Teruo Okano, Ph.D. – Research Advisor) The University of Tokyo, ( and tissue engineering research) April, 2009-March, 2014 Tokyo Women’s Medical University, (research student for a collaborative project) April, 2009-March, 2014 Work Experience: 2018-Present Co-founder, Arrow Immune INC. 2014-Present Co-founder, Sciencelounge LLC.

Selected Publications: (total 13 publications) * co-first author 1. Williford, JM*., Ishihara, J.*, …, Swartz, M.A. & Hubbell, J.A. Recruitment of CD103+ DCs via tumortargeted chemokine delivery enhances efficacy of checkpoint inhibitor. Science Advances, in press. 2. Katsumata K., Ishihara, J., …. & Hubbell, J.A. Targeting inflammatory sites through collagen affinity enhances the therapeutic efficacy of anti-inflammatory antibodies. Science Advances, in press. 3. Sasaki, K*., Ishihara, J.*, … & Hubbell, J.A. Engineered collagen-binding serum albumin as a drugconjugate carrier for cancer therapy. Science Advances, 5, eaaw6081, 2019. 4. Ishihara, J., Ishihara, A., ... Lutolf, M.P. Randi, A. & Hubbell, J.A. The heparin binding domain of von Willebrand factor functions as reservoir to promote angiogenesis in wound healing. Blood, 133, (24):2559-2569, 2019. Selected to Plenary Paper 5. Ishihara, J.*, Ishihara, A.*, … Kron, S., Swartz, M.A. & Hubbell, J.A. Targeted antibody and cytokine cancer immunotherapies through collagen affinity. Science Translational Medicine, 11, eaau3259, 2019. 6. Ishihara, J., Ishihara, A., … Swartz, M.A. & Hubbell, J.A. Improving efficacy and safety of agonistic anti- CD40 antibody through extracellular matrix affinity. Molecular Cancer Therapeutics, 17, (11):2399-2411, 2018. 7. Ishihara, J., Ishihara, A., … & Hubbell, J.A. Laminin heparin-binding peptides promiscuously bind growth factors and enhance diabetic wound healing. Nature Communications, 9(1):2168, 2018. 8. Ishihara, J., Fukunaga, K., I… Hubbell, J.A. Matrix-binding checkpoint immunotherapies enhance antitumor efficacy and reduce adverse events. Science Translational Medicine, 9, eaan0401, 2017. 9. Kumashiro, Y., Ishihara, J., … & Okano, T. Stripe-patterned thermo-responsive cell culture dish for cell separation without cell labeling. Small, 11, 681-687, 2015. 10. Ishihara, J., ... & Okano, T. Nov/CCN3 regulates long-term repopulating activity of murine hematopoietic stem cells via integrin alphavbeta3. International Journal of Hematology, 99, 393-406, 2014.

Awards/Honors: 2019 Selected as a Leading Initiative for Excellent Young Researchers (LEADER) fellow 2019 15th Annual PEGS (protein engineering summit) Boston, Selected to Poster Highlights 2019 UJA-IJC Outstanding Paper Award 2013-2015 The Japan Society for the Promotion of Science (JSPS), Fellowship 2014 Excellent Prize at The NRI Student Essay Contest, Nomura Research Institute, Ltd. 2012-2013 The University of Tokyo Global Center of Excellence, Fellowship 2011 Abstract Achievement Award, 53rd American Society of Hematology Annual Meeting MOHAMMAD ARIFUL ISLAM, PhD 1Harvard Medical School and Brigham & Women's Hospital, Boston, Massachusetts, 02115, 2Immunomic Therapeutics, Inc., Rockville, Maryland, 20850 . [email protected]

Research Overview: My research at Harvard Medical School and Brigham & Women’s Hospital was focused on developing RNA-based cancer therapeutics using nanotechnology. During this postdoctoral research I discovered a new approach to target a tumor suppressor protein, PTEN, that causes cancer progression and metastasis when it is lost in cells. I developed a PTEN mRNA tumor suppressorbased nanotherapeutic to inhibit tumor growth in various preclinical models of prostate cancer. My goal was to reintroduce functional copies of PTEN into cancer cells to turn back on the body's natural tumor suppressing mechanisms and make the tumor cells behave more like normal cells and thus be less likely to grow out of control. This technology offers a new strategy for cancer treatment and has the potential to change the currently available therapeutic options, such as target inhibitors. This discovery has been recently published in Nature Biomedical Engineering and has significant clinical relevance as comprehensive genomic sequencing of clinical specimens suggests an enormous need for restoration of tumor-suppressing pathways including PTEN. In another project, I developed a strategy to achieve robust antigenic epitope expression on immune cells to target tumor cells by delivering mRNA-antigen (or peptide-based antigen) coupled with chemically-modified adjuvant, R848. This treatment resulted in strong immunorecognition of aggtresive EG 7 lymphoma and RM1 prostate cancders, and markedly prevented or suppressed tumor growth in mice, demonstrating its potential as a potent cancer immunotherapy platform. During my doctoral work, I developed versatile polysorbitol-based nanoparticles, the first of their kind, for the delivery of oligonucleotides (DNA/siRNA/microRNA) and subunit antigens to organ sites in vivo including tumors. In my current position at Immunomic Therapeutics, Inc. I have been working on developing DNA and selfamplifying RNA-based nanovaccine formulations for cancer immunotherapy. All of these experiences have laid the foundation of my long-term goals and future research to explore: (i) combination therapeutic development upregulating tumor suppressor activity together with a down-regulation of oncogenes to boast tumor regression capacity, (ii) tumor suppressor and vaccine co-delivery with adjuvants to accelerate anti-tumor efficacy, and (iii) RNA (or self-amplifying RNA) vaccine and check-point inhibitor co- delivery to target tumor microenvironments and potentiate immunotherapeutic effects.

Education: Seoul National University, February 2013. PhD, Animal Biotechnology Seoul National University, February 2010. MS, Animal Biotechnology University of Development Alternative, September 2007. BS, Biotechnology and

Research/Work Experience: •Immunomic Therapeutics, 2018 – present Scientist, Research and Development division •Harvard Medical School and Brigham & Women’s Hospital (BWH), 2014 – 2018 Postdoctoral Research Fellow, Center for Nanomedicine; Advisors: Jinjun Shi, PhD; Bruce Zetter, PhD and Omid Farokhzad, MD •Seoul National University, 2013 – 2014 Postdoctoral Research Fellow, Center for Biomodulation, Advisors: Cheol-Heui Yun, PhD and Chong Su Cho, PhD

Selected Publications: 1.Islam MA, Xu Y, Tao W, Ubellacker JM, Lim M, Aum D, Lee GY, Zhou K, Zope H, Yu M, Cao W, Oswald JT, Dinarvand M, Mahmoudi M, Langer R, Kantoff PW, Farokhzad OC, Zetter BR, Shi J. Restoration of tumor suppression in vivo by systemic delivery of chemically modified PTEN mRNA nanoparticles. Nature Biomedical Engineering. 2: 850 (2018) 2.Islam MA, Kim S, Firdous J, Park TE, Choi YJ, Yun CH, Cho CS, Cho MH. A high affinity Kidney targeting by chitobionic acidconjugated polysorbitol transporter alleviates unilateral ureteral obstruction in rat. Biomaterials. 102:43-57 (2016) 3.Islam MA, Shin JY, Cho CS, Seo HW, Chae CH, Cho MH. The effect of RNAi silencing of p62 using an osmotic polysorbitol transporter on autophagy and tumorigenesis in lungs of K-rasLA1 mice. Biomaterials. 35:1584-96 (2014). 4.Islam MA, Shin JY, Firdous J, Park TE, Choi YJ, Cho MH, Yun CH, Cho CS. The role of osmotic polysorbitol-based transporter in RNAi silencing via caveolae-mediated endocytosis and COX-2 expression. Biomaterials. 33:8868-80 (2012). 5.Islam MA, Yun CH, Choi YJ, Shin JY, Arote R, Jiang HL, Kang SK, Nah JW, Park IK, Cho MH, Cho CS. Accelerated gene transfer through a polysorbitol-based transporter mechanism. Biomaterials. 32:9908-24 (2011).

Awards/Honors: Postdoctoral Fellowship. NIH. 2014 – 2018 Discover Brigham Research Excellence Award. BWH/Harvard Medical School. 2016 Patent Recognition Award. BWH/Harvard Medical School. 2017 Brain Korea (BK) 21 Award & Best Young Scientist Award. Seoul National University. 2012/2013 BRIAN P. JOHNSON, PhD Biomedical Engineering, UW-Madison, 1111 Highland Ave Rm6028 WIMR, Madison, WI, 53705 [email protected]

Research Overview: I have pursued interdisciplinary training in biomedical engineering, toxicology and cancer biology and developed expertise in building practical high-throughput disease models for drug discovery and toxicity testing. My research bridges the gap in biological complexity between traditional animal testing models and modern high-throughput screening approaches. To fill this void, I engineer multi-cellular culture models that reconstruct paracrine and endocrine signaling interactions for hypothesis testing and chemical screening using a manufacturing technique I developed dubbed microplate micromilling. Through this approach, I design, construct and test practical multi-culture platforms that balance biological complexity and experimental tractability. I’ve constructed models of human orofacial development and steroid signaling (funded as a K99/ROO) and prostate precision medicine (processed 26 of 30 patients in a DOD clinical trial). As evidence of their translational utility, I’ve been awarded both state (UW-SEED) and federal grants (Phase1 SBIR) to commercially develop one of these devices. These new tools are allowing me to pursue fundamental questions such as: How do chemical exposures affect morphogenic Hedgehog gradients? and What pro-agonists are we missing in modern endocrine disruptor screening assays? and have already yielded new insights into how patient specific microenvironments differentially confer chemotherapeutic resistance that would have been difficult or impossible with traditional approaches. The long-term goals of this research are to inform personalized cancer treatments and prevent disease in at-risk human populations.

Education: Postdoctoral: University of Wisconsin – Madison, Biomedical Engineering Advisor: David Beebe, (2014-present) Ph.D. University of Wisconsin - Madison, Molecular and Environmental Toxicology (2007-2013) Thesis: PAS Sensors and Their Role in Vascular Development and Drug Metabolism. Advisor: Christopher A. Bradfield B.S. Michigan Technological University, Major: Biology (Pre-Health Studies) Minor: Ecology (2004)

Research/Work Experience: • Assistant Scientist/K99 Fellow: University of Wisconsin College of Engineering, Madison, WI. (2017-present) • Chief Scientific Officer: Onexio Biosystems LLC, Madison WI (2016-present) • Postdoctoral Fellow: University of Wisconsin College of Engineering, Madison, WI. (2014-2016) • Research Assistant: McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI. (2007-2013)

Selected Publications: • Morgan, MM, Arendt LM, Alarid ET, Beebe DJ* , Johnson BP* (2019) Mammary adipose stromal cells derived from obese women reduce sensitivity to the aromatase inhibitor anastrazole in an organotypic breast model. FASEB * corresponding • Johnson BP, Ross A. Vitek RA, Huang W, Jarrard DF, Lang JM, Beebe DJ (2018) Vital ex-vivo tissue labeling and pathology guided micropunching to characterize cellular heterogeneity in the tissue microenvironment. BioTechniques, Vol 64, No. 1, 13– 19. • Morgan MM, Livingston MK, Warrick JW, Stanek EM, Alarid ET, Beebe DJ*, Johnson BP*. (2018) Mammary fibroblasts reduce apoptosis and speed estrogen-induced hyperplasia in an organotypic MCF7-derived duct model. Scientific Reports. * corresponding • Johnson, BP, Jimenez J, Beebe DJ US Patent Application No.: 62/359,977 Microtiter Plate and Uses Thereof (2016) • Morgan MM*, Johnson BP*, Livingston MK, Schuler LA, Alarid ET, Sung KE, Beebe DJ (2016) Personalized in vitro cancer models to predict therapeutic response: Challenges and a framework for improvement Pharmacology & Therapeutics, *co-first authors • Johnson BP, Walisser JA, Liu Y, Shen AL, McDearmon EL, Moran SM, McIntosh BE, Vollrath AL, Schook AC, Takahashi JS, and Bradfield CA (2014) Hepatocyte circadian clock controls acetaminophen bioactivation through NADPH-cytochrome P450 oxidoreductase. Proc Natl Acad Sci U S A.

Awards/Honors: • NIH NIEHS K99/R00 Pathway to Independence Award. 1K99-ES028744. ($946,572 direct) Sept 2018-present • Innovation AveNEW Award Society of Lab Automation-Europe, Onexio Biosystems (2019) • SBIR Advance Commercial Development Program supplement for SBIR awardees (2019, $75,000) • NIH NIEHS Small Business Innovation Research Award R43-ES029864 Role: PI (transferred) ($225,000 total) Aug 2018- present • Wisconsin State Economic Engagement and Development (SEED) Award Role: PI ($150,000 total) 2017-2018 • NIH & EPA Transform Tox Testing Challenge Finalist Role: PI ($110,000 prizes) Nov. 2017, ongoing • Best Poster Awards – Soc. of Toxicology 2013 & 2017, Teratology Soc. 2017, Gordon Conference 2015 ALASTAIR KHODABUKUS, PhD Duke University, 100 Science Drive, CIEMAS 1201A, Durham, NC, 27708 . [email protected] Research Overview: During my research career, I have developed extensive expertise in the fabrication of highly functional engineered muscle tissues derived from human iPSCs, primary myoblasts (human, rat and mouse) and immortalized human and mouse myoblasts. During my PhD and postdoc in the laboratory of Keith Baar, my research focused on the role of electrical stimulation, media conditions and satellite cell origin on engineered muscle tissue fiber-type. This work culminated in the generation of the first slow-fiber-type muscle in vitro, as assessed by myosin heavy chain (MHC) protein isoform profile, and that MHC isoform switching was dependent upon contraction length and not the commonly assumed work:rest ratio. During my postdoctoral work with Nenad Bursac, I have systematically improved the structure, function and maturation of human primary and iPSC-derived muscle tissues to generate tissues with the highest reported absolute and specific forces. I have further improved the functionality of these tissues by utilizing clinicallyrelevant xenogenic and serum-free conditions. Additionally, I have also developed functional in vitro disease models of Duchenne Muscular dystrophy and dysferlinopathy that replicate clinical phenotypes and display the most severe histopathological features reported to date for in vitro models of these rare muscle diseases. My career research goals are to identify novel molecular mechanisms underlying muscle reprogramming/specification, muscle development and muscle growth via multi-omic characterization of postnatal muscle growth in animal models and patient samples. I then plan to apply these findings to develop translationally- applicable differentiation protocols and culture conditions for the derivation of muscle progenitor cells from iPSCs, the generation of highly functional 3D tissue engineered human muscles for cellular therapies and disease modeling and the study of human muscle development in vitro.

Education: 2011 PhD, Department of Molecular Physiology, University of Dundee. 2006 BSc (Hons), 1st class honours, Sports Biomedicine, University of Dundee.

Research/Work Experience: 2015 - Present Postdoctoral Fellow, Duke University (Mentor: Nenad Bursac) 2013 - 2015 Home carer and freelance scientific writer 2011 - 2013 Postdoctoral Fellow, UC Davis (Mentor: Keith Baar) 2006 - 2011 PhD Student, University of Dundee (Mentor: Keith Baar)

Selected Publications: 1. Khodabukus A, Madden L, Prabhu N, Koves TR, Jackman CP, Muoio DM, Bursac N. (2019) Electrical stimulation increases hypertrophy and metabolic flux in tissue-engineered human skeletal muscle. Biomaterials 198:259-269. 2.Wang J*, Khodabukus, A*, Rao L, Vandusen K, Abutaleb N, Bursac N. (2019) Engineered skeletal muscles for disease modeling and drug discovery. Biomaterials. 221:119416 (*Equal Contribution) 3.Rao L, Qian Y, Khodabukus A, Ribar T, Bursac N. (2018) Engineering human pluripotent stem cells into a functional skeletal muscle tissue. Nature communications 9 (1), 126 4. Khodabukus A, Baehr LM, Bodine SC, Baar, K. (2015) Role of contraction in inducing a slow phenotype via electrical stimulationin engineered muscle. Journal of Cellular Physiology 230(10):2489-97 5. Khodabukus A, Baar, K. (2014) The effect of FBS origin on engineered skeletal muscle function, phenotype and development. Journal of Cellular Biochemistry. 115(12):2198-207 6. Khodabukus A, Baar, K. (2014) Contractile and metabolic properties of engineered muscle derived from slow and fast phenotype mouse muscle. Journal of Cellular Physiology 230(10):2489-97

Awards/Honors: 2019 - Engineering Biology for Medicine conference travel award 2016 - Present Jain Foundation Research Agreement 2011 - 2013 CIRM Stem Cell Postdoctoral Scholarship 2007 - 2011 BBSRC PhD Fellowship 2006 - EPSRC Summer Fellowship MEHMET HAMDI KURAL, PhD Yale University, Department of Anesthesiology, 10 Amistad Street, New Haven, CT, 06519 [email protected]

Research Overview: My postdoctoral work at Yale School of Medicine has focused broadly on the phenotype and function of vascular cells that are involved in atherosclerotic disorders, with an emphasis on developing treatment modalities that can alter those phenotypes and improve cardiovascular outcomes. I have two active, ongoing projects in this domain. The first project focuses on development of a novel drug-eluting stent with a unique differential effect on vascular smooth muscle and endothelial cells to prevent in-stent stenosis. I have discovered a drug combination to control smooth muscle proliferation without affecting endothelial restoration, and proved the concept with the stents that I tested in an ex vivo pig coronary artery model. Currently, I am leading the in vivo studies where we test the Fas ligand and Nitric Oxide-releasing stents in a rabbit iliac artery model. The second project is focused on potential endothelial cell therapies to control smooth phenotype in tissue engineered or autologous bypass grafts to prevent graft failure. I study the interaction between endothelial and smooth muscle cells, and vessel wall remodeling by seeding endothelial cells with different phenotypes into the lumens of freshly isolated arteries, and culturing these vessels in perfusion bioreactors. The research I would like to pursue as an independent faculty would involve 1) Creating a computational model to determine the optimal amount of the Fas ligand and nitric oxide delivered to an arterial wall after stent deployment, 2) Bioabsorbable and biologically selective drug-eluting stents, and 3) Sox-17-overexpressing ipsc-derived endothelial cell transplantation in by-pass grafts or post-percutaneous intervention. The scope spans biomedical engineering, vascular biology, materials engineering, drug delivery and has a particular emphasis on translational medicine.

Education: Doctor of Philosophy in Biomedical Engineering, 2014, Worcester Polytechnic Institute (WPI), MA Master of Science in Mechanical Engineering, 2010, Southern Illinois University Edwardsville (SIUE), IL Master of Science in Physics, 2007, Gazi University Ankara, Turkey Master of Science in Secondary Sci. and Math. Education, 2006, Middle East Technical University, Ankara, Turkey Bachelor of Science in Physics, 1999, Middle East Technical University, Ankara, Turkey

Research/Work Experience: Associate Research Scientist, Postdoctoral Associate, Yale University, Department of Anesthesiology, 2014- Present Graduate Research Assistant, Worcester Polytechnic Institute, Biomedical Engineering Department, 2010-2014 Graduate Research Assistant, Southern Illinois University Edwardsville, Mechanical Eng. Department, 2008-2010 Physics Teacher, SOBCPL High School, Nallihan, Ankara, Turkey 2000-2008

Selected Publications: 1. Kural MH*, Wang J, Gui L, Li G, Yuan Y, Leiby KL, Quijano E, Tellides G, Saltzman WM and Niklason LE, “Fas Ligand and Nitric Oxide Combination to Control Smooth Muscle Growth while Sparing Endothelium”, Biomaterials, 2019, 212:28-38 (*:corresponding author). 2 .Cakir B, Xiang Y, Kural MH, Parent M, Chapeton K, He C, Raredon MSB, Dengelegi J, Patterson B, Tanaka Y, Kim K, Sun P, Lee S., Patra P, Hyder F, Niklason LE, Yoon Y, and Park I. “Development of human brain organoids with functional endothelial cells”, Nature Methods, 2019, In Press. 3. Kural MH, Dai G, Niklason LE, Gui L, “An ex vivo vessel injury model to study remodeling” Cell Transplantation, 2018, 27(9):1375-1389. 4. Kural MH and Billiar KL, “Myofibroblast persistence with real-time changes in boundary stiffness”, Acta Biomaterialia. 2015. S1742-7061(15)30268-3. 5. Kural MH and Billiar KL, “Mechanoregulation of valvular interstitial cell phenotype in the third dimension”, Biomaterials, 2014, 35(4), 1128-1137.

Awards/Honors: R01 award from NIH (1R01HL148819-01), Budget: $2,700,000, Role: Co-investigator ($800,000 subcontract). American Heart Association Postdoctoral Fellowship, (19POST34381048), Budget: $106,000, Role: PI JAE KYOO LEE, PhD Department of Chemistry, Stanford University, Stanford, CA, 94305 . [email protected]

Research Overview: My research at Stanford University focuses on the biomedical application of water microdroplets chemistry. I have developed a new chemistry technique named microdroplet chemistry where the nature of chemical reactions occurring in micron-sized water droplets (microdroplets) is studied. These includes accelerated kinetics, altered thermodynamics, spontaneous redox reactions, nanostructure formation from metal ions, and water oxidation. The microdroplet chemistry provide new opportunities of monitoring unique bimolecular behaviors and reactions in confined environments that is similar to cells. I found that biochemical reactions including protein unfolding, protein-ligand interaction, and chlorophyll demetallation in photosynthesis chlorophylls are markedly accelerated by the factor of 1,000 or higher in microdroplets (PNAS, 2015; QRB, 2015; QRB, 2017). This microdroplet environment also enables the enzyme-free synthesis of biomolecules such as RNAs which is thermodynamically unfavorable in bulk solution (PNAS, 2017). In water microdroplets, nano-crystallization process can also spontaneously occur without any reducing agent or template (Nat Commun, 2018). I have also found that heterogeneous interface of microdroplets induces reduction of biomolecules that play significant roles in photosynthesis, respiration, and biological energy transfer (JACS, 2019). In addition, water molecules in water microdroplets are highly electrochemically active to form reactive oxygen species including hydrogen peroxide, hydroxyl radical, and superoxide (PNAS, 2019). As a biomedical application, the reactive oxygen species generated in pure water microdroplets is used for green, safe disinfection of bacterial pathogens. With these motivations, my future research will focus on the application of the findings from microdroplet chemistry to solving various biomedical problems including (1) the studies of the origin of neurodegenerative diseases such as Alzheimer's disease, (2) the studies of enzyme-free biological synthesis and metabolism, and (3) bioenergy production using spontaneous redox reactions on cellular membrane.

Education: · University of Southern California, PhD, Biomedical Engineering, 2011 · Seoul National University, MS, Biomedical Engineering, 2005 · Seoul National University, BS, Physics, 2003

Research/Work Experience: · Research Scientist, Department of Chemistry, Stanford University, 2017-Present · Postdoctoral scholar, Department of Chemistry, Stanford University, 2012-2017

Selected Publications: · Jae Kyoo Lee*, Katherine L. Walker*, Hyun Soo Han, Jooyoun Kang, Fritz B. Prinz, Robert M. Waymouth, Hong Gil Nam, Richard N. Zare. “Spontaneous Generation of Hydrogen Peroxide from Aqueous Microdroplets”. Proc Natl Acad Sci U S A., in press (* authors with equal contributions). · Jae Kyoo Lee, Devleena Samanta, Hong Gil Nam, and Richard N Zare. 2019. “Micrometer-Sized Water Droplets Induce Spontaneous Reduction”, J Am Chem Soc, 141 (27): 10585-89, 2019. · Jae Kyoo Lee, Devleena Samanta, Hong Gil Nam, Richard N. Zare, "Spontaneous formation of gold nanostructures in aqueous microdroplets", Nat Commun, 9 (1), 1562, 2018. · Jae Kyoo Lee, Hong Gil Nam, and Richard N. Zare, “Microdroplet Fusion Mass Spectrometry: Accelerated Kinetics of Acid- Induced Chlorophyll Demetallation”, Q Rev Biophys 50, e2, 2017. · Jae Kyoo Lee, Erik T. Jansson, Hong Gil Nam, and Richard N. Zare, “High-Resolution Live-Cell Imaging and Analysis by Laser Desorption/Ionization Droplet Delivery Mass Spectrometry”, Anal Chem 88(10):5453-61, 2016. · Jae Kyoo Lee*, Shibdas Banerjee*, Hong Gil Nam, Richard N. Zare, “Acceleration of Reaction in Charged Microdroplets”, Q Rev Biophys 48(4): 437–444, 2015 (* authors with equal contributions). · Jae Kyoo Lee, Samuel Kim, Hong Gil Nam, Richard N. Zare, “Microdroplet Fusion Mass Spectrometry for Fast Reaction Kinetics”, Proc Natl Acad Sci U S A 112(13):3898-3903, 2015.

Awards/Honors: · Young Investigator Award, Inaugural conference of the American Society for Nanomedicine (ASNM). · Best Poster Award, 13th Annual Fed S. Grodins Graduate Research Symposium. · Best Paper Award, Nano Bioelectronics & Systems Research Center Workshop. KYUWAN LEE, PhD Anatomy and Structural Biology, Albert Einstein College of Med, 1300 Morris Park Ave, Golding 601, BRONX, NY, 10461 . [email protected]

Research Overview: 1.MRI Imaging of Gene Expression in Live Mice - Quantification of metastasis-suppressing gene expression - Tracking of breast cancer metastasis depending on the ZBP1 expression 2. Quantitative Single-Molecule mRNA Tracking in Living Single Cells - High-Speed dark-field hyperspectral imaging Instrumentation for single molecule mRNA tracking - Ratiometric histogram for monitoring of enzyme kinetics 3. Genetically Encoded Nano-antenna Sensors - Biocompatible MRI contrast agents targeting tumors. - Genetically encoded magnetic nanoparticle probe targeting MS2-mRNA in live cells and animals - Genetically encoded TEVp-Gal complementation probe targeting MS2-PP7-mRNA in live cells and animals - Microinjected gold nanoparticle dimers targeting single alternative splicing of mRNA in live cells - Gold nanoparticle assemblies targeting single cell surface markers. 4. Surface Plasmon Theory and Simulation - Spectral intensity change, broadening, and peak shift of nano-antenna dimers and multimer structures by materials, shapes, sizes, structures - Photothermal heating, assembly and disassembly control of nanoparticle structures by heating

Education: Ph.D., 2011, Purdue University, Biological Engineering, West Lafayette, IN MS, 2007, University of Pittsburgh, Physics, Pittsburgh, PA BS, 2004, Korea University, Physics (Summa Cum-Laude), Seoul, Korea

Research/Work Experience: 1. Albert Einstein College of Medicine, Research Fellow (Mentor: Robert H. Singer) 2016- 2. Intel Corp, PTD R&D Engineer 2012-2016 3. University of California Berkeley, Postdoctoral Scholar (Mentor: Luke P. Lee) 2011-2012 4. Internship, Samsung Advanced Institute of Technology, Korea 2010 5. Purdue University, PhD Student (Adviser: Joseph Irudayaraj) 2007-2011

Selected Publications: 1. Kyuwan Lee, Robert H. Singer*. “Single Molecule Tracking in Live Cells: Direct Comparison of Energy Transfer Pairs and Plasmonic Dimers for Hyperspectral Imaging”, Trend in Biotechnology (Invited), 2019, In Revision 2. Kyuwan Lee, Yi Cui, Luke P. Lee*, and Joseph Irudayaraj*. “Quantitative Imaging of Single mRNA Splice Variants in Living Cells”, Nature Nanotechnology, 2014, 9, 474-480 3. Kyuwan Lee and Joseph Irudayaraj*. “Correct Spectral Conversion between Surface-enhanced Raman and Plasmon Resonance Scattering from Nanoparticle Dimers for Single Molecule Detection”, Small, 2013, 9, 1106-1115 4. Kyuwan Lee, Vladimir Drachev, and Joseph Irudayaraj*. “DNA-Gold Nanoparticle Networks Grown at Cell Surface Marker Sites: Application in Diagnostics”, ACS Nano, 2011, 5, 2109-2117 5. Kyuwan Lee and Joseph Irudayaraj*. “Periodic and Dynamic 3-D Gold Nanoparticle-DNA Network Structures for Surface- Enhanced Raman Spectroscopy-Based Quantification”, The Journal of Physical Chemistry C, 2009, 113, (15), 5980-5983

Awards/Honors: 1.KSEA Excellent Presentation Award 2.Deans Scholarship, University of Pittsburgh, Physics 3.Highest Honor, Korea University, Physics 4.Baekwoon Award and Gold Medal, Korea University, Physics 5.Kwanjoung Education Foundation Award and Scholarship 6.Honor Scholarship, Korea University, Physics PAUL (SEUNG YUP) LEE, PhD Biomedical Engineering, Georgia Tech/Emory University, 1760 Haygood Dr. NE, Atlanta, GA, 30322 [email protected]

Research Overview: My long-term research goal is to develop and translate the next generation of optical sensing technology for rapid, quantitative, and noninvasive assessment of biological tissues. I am particularly interested in miniaturized optoelectronic sensors on a wearable platform coupled with computational (including machine learning) algorithms to quantify biophysically-relevant parameters. From my research experience, potential applications include blood flow and tissue oxygenation monitoring for pediatric brain diseases, pancreatic cancer diagnosis, and tissue viability monitoring. My PhD studies at Michigan focused on diffuse reflectance and fluorescence spectroscopy for pancreatic cancer detection and viability assessment of implanted tissue-engineered constructs. Extending my research as a postdoctoral fellow at Michigan, I designed and fabricated a needle-compatible ultra-miniaturized optoelectronics sensor to probe human pancreatic tissues in situ. I also validated a compact diffuse optics system to monitor perfusion of a transferred skin flap. Afterwards, I continued to work as a postdoctoral fellow at Georgia Tech/Emory University where I am focusing on multiple approaches for translational diffuse optics for pediatric brain monitoring. First, I employ diffuse correlation spectroscopy to measure microvascular cerebral blood flow in children with sickle cell disease at the Children’s Healthcare of Atlanta. Secondly, I am developing a miniaturized speckle contrast optical spectroscopy system to estimate tissue oxygen metabolism. My strong electrical/optical/biomedical engineering background and versatile clinical and translational research experience has established the foundation for a highly multidisciplinary research program to pursue my overarching goal of making noninvasive optical sensing ubiquitous for personalized medicine.

Education: • Ph.D. Biomedical Engineering 2015, University of Michigan, Ann Arbor, MI • M.S. Biomedical Engineering 2006, Seoul National University, South Korea • B.S. Electrical Engineering 2004, Seoul National University, South Korea

Research/Work Experience: • Postdoctoral Fellow, 2016 – present, Department of Biomedical Engineering at Georgia Tech/Emory Univ. • Postdoctoral Fellow, 2015 – 2016, Department of Biomedical Engineering at University of Michigan • Graduate Student Instructor, 2013 – 2014, Year-long BME design class, University of Michigan • Graduate Research Fellow, 2011 – 2015, Department of Biomedical Engineering at University of Michigan • Research Engineer, 2006 – 2010, Korea Electrotechnology Research Institute, South Korea

Selected Publications: • Lee, S.Y, Cowdrick, K., Sanders, B., Sathialingam, E., Lam, W., Joiner, C.H., Buckley, E. M., " Noninvasive optical assessment of resting-state cerebral blood flow in children with sickle cell disease”, Neurophotonics; 2019 6(3): 035006 • Lee, S.Y., and Mycek, M.-A., “Hybrid Monte Carlo simulation with ray tracing for fluorescence measurements in turbid media” Optics Letters, 2018; 43(16): 3846-3849 • Lee, S.Y., Chung, G. Y., and Mycek, M.-A., “In situ optical tissue diagnostics/Miniaturized optoelectronic sensors for tissue diagnostics”, Encyclopedia of Modern Optics 2nd edition, Oxford Elsevier, 2018; Vol 3:86-94 • Lee, S.Y*., Pakela J. M*. et. al., “Compact dual-mode diffuse optical system for blood perfusion monitoring in a porcine model of microvascular tissue flaps”, Journal of Biomedical Optics, 2017; 22(12): 121609 *(Equal contribution) • Lee, S.Y., Lloyd, W.R., Chandra, M., Wilson, R.H., McKenna, B., Simeone, D., Scheiman, J., and Mycek, M.-A., "Characterizing human pancreatic cancer precursor using quantitative tissue optical spectroscopy," Biomedical Optics Express, 2013; 4(12):2828-34

Awards/Honors: American Heart Association Postdoctoral Fellowship, 2019 – 2020 | Best Talk Award (1st place), 2018 5th Georgia Tech Postdoc Research Symposium | Best Scientific Poster Award (2nd place), 2018 Southeastern Pediatric Research Conference | Best Seminar Award, 2018 Department of Biomedical Engineering, Georgia Tech/Emory Univ. | Petit Scholar Mentor, 2017 & 2018, Petit Institute of Biological Engineering, Georgia Tech | Rackham Centennial Fellowship, 2013 University of Michigan. | Department Graduate Fellowship, 2010 University of Michigan WONJAE LEE, PhD Stanford University, 300 Pasteur dr. R-201, Stanford, CA, 94305 . [email protected]

Research Overview: My research goal is to engineer experimental tissue platforms to address the dynamics of individual neural cell behaviors and illuminate the dynamic geno-pheno-envirotype relationship. I approach this goal by reconstructing functional neurovascular unit (NVU), consisting of microfluidic vasculature and the neighboring engineered 3D brain tissue, and investigating the cytoskeletal behaviors of the cells around it. My background in the mechanical engineering is an important asset in rebuilding the hierarchical NVU micro-structure, simulating biomechanical conditions by the blood flow, and analyzing how cells rearrange cytoskeletal networks to generate mechanical force. And my research training in tissue engineering laid a firm technical foundation to build functional NVU microenvironment in both healthy and pathological conditions.

Education: Ph.D., Mechanical engineering, Stanford University, U.S.A. (01/2007 – 04/2010) M.S., Biomechanical engineering, Stanford University, U.S.A. (09/2002 – 12/2006) B.S., Mechanical engineering, POSTECH, S. Korea, (03/1997 – 02/2001)

Research/Work Experience: Instructor at Stanford University (09/2013 – present) - Development of microfluidic vasculature models · restore the structural and functional integrity of various human tissues around vasculature in microfluidic chips - Patterned stem cell differentiation in 3D hydrogel matrices · control physicochemical stimuli temporospatially in 3D hydrogel matrices for reconstructing heterocellular tissue structures Postdoctoral fellow at Stanford University (04/2010 – 08/2013) • Supervised by Prof. Jon Park (Neurosurgery) - Axon extension guidance · established an axon guiding matrix by generating a linear propagation of the axon attractant releases - In vitro experimental model of cancer angiogenesis · constructed vascularized engineered tumors using microfabricated structures Graduate student researcher at Stanford University • Supervised by Prof. Curtis Frank (Chemical engineering) and Prof. Jeffrey Glenn (Medicine) (01/2007 - 04/2010) - Hydrogel based liver tissue engineering (Ph.D. dissertation topic) · modulated the physical properties of the hydrogel network by incorporating hydrophobic nanoparticles · controlled drug-releasing rate from poly(lactic-co-glycolic acid) microparticles · constructed microstructure of the liver lobule using microfluidics and soft lithography • Supervised by Prof. Daniel Kim (Neurosurgery) (09/2004 – 06/2006) - Design of spinal prostheses for noninvasive surgery · analyzed mechanical loading using computational methods (MATLAB and ANSYS) · treated the surface of prostheses for tissue integration • Supervised by Prof. Lane Smith (Orthopedic Surgery) (06/2003 – 04/2004) - Analysis of osteogenic stimulation by applying shear stress

Selected Publications: - W. Lee, C.W. Frank, J. Park, Directed Axonal Outgrowth Using a Propagating Gradient of IGF-1, Adv. Mat., 2014, 26, 4936-4940. - W. Lee, J. Park, Design of heterocellular 3D architecture and its application to monitoring the behavior of cancer cells in response to the spatial distribution of endothelial cells, Adv. Mat., 2012, 24, 5339-5344. - W. Lee, N.J. Cho, A. Xiong, J.S. Glenn, C.W. Frank, Hydrophobic nanoparticles improve permeability of cell- encapsulating poly(ethylene glycol) hydrogels while maintaining patternability, Proc. Natl. Acad. Sci., 2010, 107, 20709- 20714.

Awards/Honors: - Career Development Award (K25, NIH) 01/2017 – 12/2021 - Mogam Scientific Scholarship (Mogam Foundation, S. Korea) 09/2006 SASHA CAI LESHER-PÉREZ, PhD Marie-Curie Individual Fellow, Elvesys SAS, 83 Avenue Philippe Auguste, Paris, France 75011 [email protected]

Research Overview: My research has focused on developing micro-engineered tools to manipulate the cellular microenvironment, in order to recapitulate and analyze tissue- and organ-level function and pathophysiological phenomena. My goals are to build surrogate biological models on-chip (tissue- and organ-on-chip technologies), which will enhance the evaluation of human disease progression and intervention at the molecular, cellular, and organ-level, as complementary toolsets with in vivo animal models. During my doctoral research, I developed self-regulating microfluidic circuits for the temporal control and manipulation of the on-chip fluidic environment, methods for enhancing the high-throughput formation of spheroids for the facile construction of three-dimensional cellular aggregates, and a versatile oxygen monitoring system for three- dimensional cell culture platforms. Engineering microsystems to build, manipulate and analyze tissue cultures, led me to my postdoctoral position at UCLA. I used microfluidic systems to build microparticle hydrogel building blocks as therapeutic biomaterials. The modular, tunable microparticles, while clinically relevant, can also be assembled into more complex tissue culture scaffolds for surrogate tissue models and provide a heterogenous mechanical and chemical milieu to better mimic the native microenvironment. While at UCLA, I received a Marie-Curie Fellowship to work with a French startup, Elvesys. In my current position, I design microfluidic devices with proprietary thermoplastics, for the facile fabrication of bio-compatible or chemo-compatible devices for higher throughput manufacturing, reduced absorption of small molecules, and material stability. These devices are primarily aimed to produce organ-on-chip and drug development platforms. I plan on building my research program around biological models that can more aptly capture the pathophysiological phenomena, specifically focusing on the dysregulation of glucocorticoids and their role in metabolic and neurological disorders and diseases.

Education: • University of Michigan, May 2015. PhD, Biomedical Engineering • University of Wisconsin - Madison, December 2009. BS, Biomedical Engineering

Research/Work Experience: • Elvesys SAS, 2018 – present Marie-Curie Individual Fellow, Microfluidic Innovation Center; CEO: Guilhem Velve-Casquillas, PhD • University of California, Los Angeles, 2015-2018 Ford Foundation Postdoc Fellow, Chemical and Biological engineering; Advisor: Tatiana Segura, PhD • University of Michigan, 2009-2015 Graduate Research Fellow, Biomedical Engineering; Advisor: Shuichi Takayama, PhD • Johnson and Johnson, 2009, 2011 GEM Fellow (2009) and CBTP Fellow (2011), Consumer Products Division; Supervisor: Carol Gell, PhD

Selected Publications: * -denotes equal contribution 1. Lesher-Pérez SC*, Zhang C*, and Takayama S. Capacitive coupling synchronizes autonomous microfluidic oscillators. Electrophoresis. 39(8): 1096-1103. (2018) 2. Lesher-Pérez SC, Kim G-A; Kuo C, Leung BM, Mong S, Kojima T, Moraes C, Thouless MD, Luker GD, and Takayama S, Dispersible oxygen microsensors map oxygen gradients in three-dimensional cell cultures. Biomaterials Science. 5(10):2106-2113. (2017) 3. Leung BM*, Lesher-Perez SC*, Matsuoka T, Moraes C, and Takayama S. Media additives to promote spheroid circularity and compactness in hanging drop platform. Biomaterials Science. 3(2): 336-344. (2014) 4. Lesher-Perez SC*, Weerappuli P*, Zhang C, Kim S-J, and Takayama S. Predictable duty cycle modulation through coupled pairing of syringes with microfluidic oscillators. Micromachines. 5(4): 1254-1269. (2014) 5. Lesher-Perez SC, Frampton JP, and Takayama S. Microfluidic systems: A new toolbox for pluripotent stem cells. Biotechnology Journal. 8(2): 180-191. (2013)

Awards/Honors: • Horizon 2020 Marie Skłodowska-Curie Individual Fellowship. European Union H2020 Program. 2018 - present • Ford Foundation Postdoctoral Fellowship. NAS. 2017-2018 • Diversity Supplement Postdoctoral Fellowship, F31 Grant. NIH/NINDS. 2015-2017 • NSF Graduate Research Fellowship. 2012-2015 DAVID S. LI, PhD Department of Chemical Engineering, University of Washington, 105 Benson Hall, Box 351750, Seattle, WA 98195-1750 Department of Bioengineering, University of Washington, Benjamin Hall Building, Rm 363, Seattle, WA 98195-1750 ([email protected])

Research Overview: My PhD research was focused on the dynamics of droplets and bubbles in the vasculature in the context of an ultrasound- based therapy called gas embolotherapy. Gas embolotherapy is the selective vaporization of perfluorocarbon microdroplets, using focused ultrasound, near a tumor site to generate gas emboli that would lodge in the tumor vasculature, redirecting blood flow to starve off the tumor. I was able to determine the mechanism of acoustic droplet vaporization and the role non-linear wave propagation has in initiating vaporization. Additionally, I studied the dynamics of bubble transport and potential secondary interactions ultrasound may have with bubbles in the vasculature using experiments and computational tools. During my PhD I became proficient in computational modeling, microfluidics, high-speed imaging, CAD, and particle imaging velocimetry. As a postdoc, my primary research is on developing a nonlinear contrast-based ultrasound imaging and therapy technique called sonophotoacoustics. The idea behind sono- photoacoustics is that nanodroplets small enough to penetrate tissue for extravascular imaging and therapy can be easily synthesized, but the optical or acoustic energies needed to vaporize the droplets to generate photoacoustic or ultrasound contrast can be prohibitively high. By using a combination of a photothermal heating from a laser pulse with the negative pressure phase of an acoustic pulse, the activation energy needed to vaporize nanodroplets can be reduced by as much as two orders of magnitude, well within FDA and ANSI limits. I have developed imaging sequences and scanning protocols for real time sonophotoacoustic contrast imaging interleaved with conventional ultrasound and photoacoustic images. Additionally, by adjusting sonophotoacoustic settings the vaporized droplets can be used for therapy, such has breaking up blood clots. Moreover, I have developed new droplet synthesis methods to consistently and easily synthesize nanodroplets under 200 nm in diameter using a spontaneous droplet nucleation.

Education: • University of Michigan, May 2014. Ph.D. Biomedical Engineering • University of Michigan, May 2008. B.S.E Biomedical Engineering

Selected Research/Work Experience: • University of Washington, Seattle, WA, 2015-Present Postdoctoral Researcher, Chemical Engineering & Bioengineering, Advisors: Lilo Pozzo, Ph.D. (Chemical Engineering) & Matthew O’Donnell, Ph.D. (Bioengineering) • University of Michigan, Ann Arbor, MI, 2008-2015 Graduate Student Research Assistant and Postdoctoral Researcher, Biomedical Engineering, Advisor: Joseph Bull, Ph.D. • Vir(Sn) Inc., Ann Arbor, MI, 2008-2013 Co-founder & Engineer

Selected Publications: 1. D.S. Li, S. Schneewind, M. Bruce, M. O’Donnell, and L.D. Pozzo. “Spontaneous Nucleation of Perfluorocarbon Emulsions”. Nano Letters. 2018, 19 (1), 171-181. 2. D.S. Li, Y.-T. Lee, J. Ilavsky, I. Kuzmenko, G.-S. Jeng, M. O’Donnell, and L.D. Pozzo. “Ultrasound Synthesis of Nano-Pickering Emulsions Investigated via In-Situ SAXS”. J. Colloid Interface Sci. 2019, 536, 281-290 3. D.S. Li, Y.-T. Lee, Y. Xi, I. Pelivanov, M. O’Donnell, and L.D. Pozzo. “A Small-Angle Scattering Environment for In Situ Ultrasound Studies”. Soft Matter. 2018. 14, 5283 4. D.S. Li, S.J. Yoon, I. Pelivanov, M. Frenz, T. Matula, M. O’Donnell, and L.D. Pozzo. “Polypyrrole-Coated Perfluorocarbon Nanoemulsions as a Sono-Photoacoustic Contrast Agent”. Nano Letters. 2017. 17(10), 6184-6194 5. D.S. Li, O.D. Kripfgans, M.L. Fabiilli, J.B. Fowlkes, and J.L. Bull, “Initial Nucleation Site Formation Due to Acoustic Droplet Vaporization”. Applied Physics Letters. 2014. 104(6), 063703 6. D.S. Li, O.D. Kripfgans, M.L. Fabiilli, J.B. Fowlkes, and J.L. Bull, “Formation of Toroidal Bubbles from Acoustic Droplet Vaporization”. Applied Physics Letters. 2014. 104(6), 063706

Selected Awards/Honors: Selected Accepted Beam Time Proposals: • “In Situ Scattering Measurements of Phase-Changing Contrast Agents”, NIST Center for Neutron Research. 2018 • “In Situ Characterization of Droplet-Based Ultrasound Contrast Agents”, Australian Synchrotron. 2018 • “Structural Analysis of Nanoemulsion for Diagnostic Imaging and Therapy”, Argonne National Lab. 2016-2018 Selected Travel Grants and Awards: Rackham Travel Fellowship – 2010-2013 GAANN Fellowship –2009-2010 & 2012-2013 Dare to Dream Grant – March 2009 JIE LI, PhD Bioengieering, University of California Berkeley, 140 Hearst Memorial Mining Building, Berkeley, CA, 94720 [email protected]

Research Overview: I work on CRISPR based genome editing using non-viral vector such as lipids and polymers for intracelullar delivery of Cas9-RNP. At City of Hope I focused on nanoparticle/ hybrids for targeted tumor therapy. Specifically, paclitaxel nanocrystal was encapsulated with thin layer of silica and loaded into neural stem cells with excellent tumor tropism for ovarian cancer treatment. At UCLA I focused on biomacromolecules delivery based on synthetic materials. Specifically, in-situ polymerization of proteins or self-assembly of proteins with polymers for intracellular delivery and drug long circulation.

Education: 2010 Bachelor of Science, Chemistry, Nankai University 2010 Bachelor of Engineering, Chemical Engineering, Tianjin University 2015 PhD, Chemical Engineering, University of California, Los Angeles

Research/Work Experience: Postdoc Fellow at University of California, Berkeley, Bioengineering Department (Current) • Developing and screening nonviral delivery vector for CRISPR-Cas9 gene editing Postdoc Fellow at Beckman Research Institute of City of Hope (2016 – 2017) • Developed nanoparticles/neural stem cell hybrids for novel targeted cancer therapy. Postdoc Researcher at Chemical and Biomolecular Engineering Department (2015 – 2016) • Developed degradable self crosslink nanocapsule for protein delivery Graduate Researcher at Chemical and Biomolecular Engineering Department (2010 – 2015) • Developed single protein nanocapsule for chemical warfare agents detoxification • Developed highly robust enzyme silica composites based on protein nanocapsules • Developed pH sensitive zwitterionic polymer protein conjugates for tumor site targeting Teaching Fellow (2011 - 2015) • Transport Phenomena, Separation Process, Chemical Reaction Engineering, Chemical Process Computer-Aided Design and Analysis, Process Economics and Analysis Reviewer for peer-reviewed journals • Chemical Communication, Chemical Science, Journal of Material Chemistry B

Selected Publications: 1. J. Li, P. Tiet, W. Abidi, R. Mooney, L. Flores, S. Aramburo, et al., Silica coated paclitaxel nanocrystals enable neural stem cell loading for treatment of ovarian cancer, Bioconjug. Chem. 2019, 30, 1415–24. 2. J. Li, J. Røise, J. Zhang, J. Yang, D. Kerr, H. Han, N. Murthy, A novel fluorescent surfactant enhances the delivery of the Cas9 ribonucleoprotein and enables the identification of edited cells, Chem. Commun., 2019, 55(31), 4562–5. 3. R. Rouet, L. de Oñate, J. Li, N. Murthy, R.C. Wilson, Engineering CRISPR-Cas9 RNA-Protein Complexes for Improved Function and Delivery, The CRISPR Journal, 2018, 1, 367–78. 4. Y. Zhang, J. Røise, K. Lee, J. Li, Recent developments in intracellular protein delivery, Curr. Opin. Biotechnol., 2018, 52, 25-31. 5. S. Liang, Y. Liu, X. Jin, G. Liu, J. Wen, L. Zhang, J. Li, et al., Phosphorylcholine polymer nanocapsules prolong the circulation time and reduce the immunogenicity of therapeutic proteins, Nano Res., 2016, 1-10. 6. J. Li, Y. Liu, J. Wen, D. Wu, D. Xu, L. Zhang, J. Jin, H. Wang, Y. Lu, An intracellular protein delivery platform based on glutathione-responsive protein nanocapsules, Chem. Commun., 2016, 52, 13608-13611. 7. J. Li, X. Jin, Y. Liu, X. Zhu, Y. Lu, Robust Enzyme-Silica Composites Made from Enzyme Nanocapsules, Chem. Commun., 2015, 51, 9628-9631. 8. J. Li, Y. Liu, Y. Lu, Enzyme therapeutics for systemic detoxification, Adv. Drug. Deliv. Rev., 2015, 90, 24–39. 9. J. Du, J. Jin, Y. Liu, J. Li, T. Tokatlian, Z. Lu, T. Segura, X. Yuan, X. Yang, Y. Lu, Gold-Nanocrystal-Enhanced Bioluminescent Nanocapsules, ACS Nano, 2014, 8, 9964-9969. 10. W. Wei, J. Du, J. Li, M. Yan, Q. Zhu, X. Jin, X. Zhu, Z. Hu, Y. Tang, Y. Lu, Construction of Robust Enzyme Nanocapsules for Effective Organophosphate Decontamination, Detoxification, and Protection, Adv. Mater., 2013, 25, 2212-2218.

Awards/Honors: National Scholarship (Ministry of Education of China) Teaching Fellow (UCLA) University Fellowship (UCLA) YAMIN LI, PhD Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA, 02155 . [email protected]

Research Overview: My research will be focused on the development of novel lipid and polymer-based materials for biomedical applications (e.g. gene delivery, protein and peptide delivery, drug delivery, gene editing, biomedcial imaging, biomarker detection, artificial enzymes, etc.). My doctoral dissertation was focued on the design and synthesis of stimuli-responsive polymerss for sensing, imaging, and antimicrobial/anticancer drug delivery. My postdoc research is focused on the development of synthetic lipid nanoparticles for intracellular delivery of proteins and genes for cancer therapy and genome editing.

Education: Doctor of Science in Chemistry, 2016, University of Science and Technology of China Bachelor of Science in Chemistry, 2011, University of Science and Technology of China

Research/Work Experience: 2016-present, Postdoctoral Scholar, Tufts University 2011-2016, Graduate Research Assistant, University of Science and Technology of China 2009-2011, Undergraduate Research Assistant, University of Science and Technology of China

Selected Publications: 1. First and co-first authored publications: ACS Biomaterials Science & Engineeing, 5, 2391 (2019); Advanced Healthcare Materials, 8, 1800996 (2019); Biomaterials Science, 7, 597 (2019); Nature Biomedical Engineering, 2, 469 (2018); Biomaterials, 178, 652 (2018); Journal of Controlled Release, 263, 39 (2017); Acta Polymerica Sinica, 7, 1178 (2017); Angewandte Chemie International Edition, 55, 1760 (2016); Advanced Materials, 26, 6734 (2014); Biomaterials, 35, 1618 (2014); Biomacromolecules, 13, 3877 (2012). 2. Co-authored publications: , 26, 187 (2019); Clinical Theriogenology, in press (2019); Nature, 553, 217 (2018); Nature Biomedical Engineering, 1, 854 (2017); Trends in Biotechnology, 36, 173 (2017). 3. Patents: US Patent (Application No.62/625,153,2018, Pending); China Patent (Application No. 201510237278.5,2015, Issued; Application No. 201510237260.5,2015, Issued; Application No. 201410205717.x,2014, Issued)

Awards/Honors: 1.Top 10 Outstanding Reviewers for Molecular Systems Design & Engineering, Royal Society of Chemistry, 2017 2.Top 10 Reviewers for Biomaterials Science, Royal Society of Chemistry, 2016 3.Best Presentation Award (First Prize), 5th Annual Meeting of Chemistry, USTC, 2016 4.Yuan Dong Scholarship Award (First Prize), USTC, 2015 5.Best Presentation Award (Second Prize), 9th Annual Meeting of Science, HFNL, 2015 6.Best Young Researcher, International Symposium on Transnational Nanomedicine, Guangzhou, 2015 7.Yuan Dong Scholarship Award (First Prize), USTC, 2014 8.Yuan Dong Scholarship Award (Second Prize), USTC, 2013 9.Best Presentation Award (Second Prize), 7th Annual Meeting of Science, HFNL, 2013 10.Best Presentation Award (First Prize), 2nd Annual Meeting of Chemistry, USTC, 2013 11.National Scholarship, Ministry of Education of China, 2012 12.Excellent Graduation Thesis, USTC, 2011 STEPHANIE E. LINDSEY, PhD Department of Pediatrics, Institute for Computational & Mathematical Engineering, Stanford University, Clark Center- E100B, Stanford, CA, 94305 . [email protected]

Research Overview: My research specializes in developmental biomechanics, with the goal of engineering new solutions for the treatment of cardiovascular malformations. Through a combination of targeted experimental manipulations and multiscale computational modeling, I work to establish quantitative relationships between hemodynamics and tissue growth and remodeling in cardiac development. The main goals of my research are to 1) To delineate the role aberrant hemodynamics plays in the creation of clinically relevant cardiovascular malformations and 2) to design the most effective interventions to reverse abnormal flow conditions. Further advances in surgical interventions require a more complete understanding of the etiology of congenital heart diseases. New experimental and computational techniques can further this understanding and enable direct prediction of mechanical perturbations, such as vessel occlusions, on cardiovascular development. Regenerative strategies can then utilize the developmental paradigms uncovered. By performing targeted manipulations and designing the computational tools necessary to optimize research findings, my future research group will be able to explore each of its experimental findings from multiple perspectives.

Education: Ph.D., Biomedical Engineering, 2016, Cornell University, Ithaca, NY M.S., Biomedical Engineering, 2013, Cornell University, Ithaca, NY B.S., Biomedical Engineering, 2010, Washington University, St. Louis, MO

Research/Work Experience: • Postdoctoral Associate (Advisor: Alison Marsden), Stanford University, Stanford, CA - Quantification and optimization of growth and remodeling in tissue engineered vascular grafts through numerical simulation •. Postdoctoral Associate (Advisors: Simon Mendez, Pascal de Santa Barbara), University of Montpellier, Montpellier, France - Development of a direct forcing fluid-structure interaction solver within the YALES2BIO highly parallel finite-volume solver - Quantification of changing gastro-intestinal tract function through ultrasound and soft tissue mechanics •. Graduate Research Associate (Advisor: Jonathan Butcher), Cornell University, Ithaca, NY - Dissertation Title: Hemodynamic Regulation of Cardiac Outflow Morphogenesis - Quantification of cardiac morphogenesis through experimental and computational techniques in the avian embryo • Graduate Research Intern (Mentor: Irene Vignon-Clementel), INRIA Paris, Rocquencourt, France - Creation of multiscale computational simulations of cardiac morphogenesis • Undergraduate Research Associate (Mentor: Larry Taber), Washington University, St. Louis, MO - Quantification of the mechanics of the developing ferret brain and the gastrulating chick embryo by way of microindentation experiments and germ-layer tracking

Selected Publications: 1. Lindsey, S.E., Butcher J.T., & Vignon-Clementel, I.E., Cohort-based multiscale analysis of hemodynamic-driven growth and remodeling of the embryonic pharyngeal arch arteries. Development. Vol 145 (20). 2018. 2. Ryvlin, J*#, Lindsey, S.E.#, Butcher J.T., Systematic analysis of the smooth muscle wall phenotype of the pharyngeal arch arteries during their reorganization into the great vessels and its association with hemodynamics. Anat. Rec., Vol 302. 2019:153-162. 3. Lindsey, S.E.#, Menon, P.G.# et al. Growth and hemodynamics after early embryonic aortic arch occlusion. Biomech Model Mechanobiol, Vol 14. 2015:735-751. 4. Lindsey, S. E., Butcher, J. T., & Yalcin, H. C. Mechanical regulation of cardiac development. Frontiers in Physiology, Vol 5. 2014. * mentored undergraduate student; # co-first authors.

Awards/Honors: 2018-2020 NIH T32 Mechanisms and Innovations in Cardiovascular Disease Postdoctoral Fellow 2015-2017 Whitaker Postdoctoral Research Scholar, International Institute of Education 2015 Federation of American Societies for Experimental Biology BioArt Competition Winner 2010-2015 Alfred P. Sloan Fellow 2013- 2014 National Science Foundation Graduate Research Opportunities Worldwide Fellow 2011-2014 National Science Foundation Graduate Student Research Fellow 2009-2010 NIH MARC Undergraduate Student Training in Academic Research Fellow CLAUDIA LOEBEL, MD, PhD Bioengineering, University of Pennsylvania, 240 Skirkanich Hall, 210 S. 33rd Street, Philadelphia, Pennsylvania, 19104 [email protected] Research Overview: My research program will be focused on elucidating the complex biological signals of the cellular microenvironment that form and maintain tissues in the body. Specifically, I aim to answer fundamental questions about how the local extracellular matrix (ECM) presents and preserves both physical and biochemical cues to direct development and regeneration of organs. My approach will include the application of technology for the fabrication of biomaterials and engineered cellular systems with the ultimate goal of addressing biological, medical and societal needs. Central to my work will be a focus on reproducing biological features at multiple scales using engineering principles towards the development and translation of more effective therapeutic treatments. With this general goal in mind, my group will pursue the folloiwing areas: (1) Fundamental understanding of the relationship between multicellular self-organization and nascent matrix biophysics and mechanotransduction, (2) the use of microengineering tools to study molecular mechanism that regulate disease and long-term memory of regenerative programs in organoid systems, and (3) the application of these experimental tools as therapeutic strategy to modulate regenerative activity as cell transplants. Although this fundamental work will be of interest across broad fields, I initially look to apply the work to regulating the cellular response to hydrogels and pulmonary disease.

Education: Ph.D. Health Sciences and Technology, October 2016, ETH Zurich, Switzerland, Advisors: Drs. Marcy and David Eglin M.D., November 2011, Martin-Luther University Halle-Wittenberg, Germany, Advisor: Dr. Frank Bartel

Research/Work Experience: Postdoctoral Fellow, University of Pennsylvania, 2016-present, Department of Bioengineering, Advisor: Dr. Jason A. Burdick - Designed a bio-orthogonal extracellular matrix labeling technique to understand the role of nascent protein secretion in the cellular perception of engineered matrices. - Developed microstructured hydrogels to produce lung epithelial organoids and determine biophysical and paracrine signaling mechanisms during lung alveolar regeneration. Graduate Student, ETH Zurich and AO Research Institute Davos, 2012-2016, Department of Health Science and Technology, Advisors: Drs. Marcy Zenobi-Wong, David Eglin Visiting Graduate Student, University of Pennsylvania, 2015, Department of Orthopaedic Surgery, Advisor: Dr. Robert L. Mauck - Investigated early markers of differentiation to predict the osteogenic potential of human mesenchymal stromal cells in vitro. - Developed a hydrogel platform to probe cell behavior in response to hydrogel crosslinking mechanisms.

Selected Publications: 1. Loebel, C.; Mauck, R.L.; and Burdick, J.A. (2019) Local nascent protein deposition and remodeling guide mesenchymal stromal cell mechanosensing and fate in three-dimensional hydrogels. Nature Materials, 18, 883-891 2. Loebel, C. and Burdick, J.A. (2018) Engineering Stem and Stromal Cell Therapies for Musculoskeletal Tissue Repair. Cell Stem Cell, 22, 325-339 3. Loebel, C.*; Rodell, C.B.*; Chen, M.H.; Burdick, J.A. (2017) Shear-thinning and self-healing hydrogels as injectable therapeutics and for 3D-printing. Nature Protocols, 12(8), 1521-1541 *equal contribution 4. Loebel, C.; Szczesny, S. E.; Cosgrove, B. D.; Alini, M.; Zenobi-Wong, M.; Mauck, R. L.; Eglin, D. (2017) Crosslinking Chemistry of Tyramine-Modified Hyaluronan Hydrogels Alters Early Attachment and Behavior. Biomacromolecules, 18(3), 855-864 5. Loebel, C.*; Broguiere, N.*; Alini, M.; Zenobi-Wong, M.; Eglin, D. (2015) Microfabrication of Photo-Cross-Linked Hyaluronan Hydrogels by Single- and Two-Photon Tyramine Oxidation. Biomacromolecules, 16(9), 2624-30 *equal contribution 6. Loebel, C.; Czekanska, E.M.; Bruderer, M.; Salzmann, G.; Alini, M.; Stoddart, M.J. (2014) In vitro osteogenic potential of human mesenchymal stem cells is predicted by Runx2/Sox9 ratio. Tissue Engineering Part A, 21(1-2), 115-23

Awards/Honors: ETH Silver Medal, 2018; Julia Polak European Doctoral Award, 2017; Research Award, Swiss Society for Biomaterials and Regenerative Medicine, 2017; Racquel Z. LeGeros Award, European Society for Biomaterials, 2015; Best Overall Oral Presentation Award, European Orthopaedic Research Society, 2014 WEI LV, PhD Chemical & Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlasnta, GA, 30318 . [email protected]

Research Overview: With expertise in biomaterial engineering, drug delivery system, microbiology, and cell biology, my research goal is to leverage protein engineering approaches to develop next-generation biomaterials and therapeutic strategies for tissue regeneration, bacterial infection and cancer therapy. My graduate research sought to develop multifunctional biomaterials that promote tissue regeneration and prevent bacterial infection, which included new rechargeable drug delivery systems and analytical methods for bacterial-cellbiomaterial interactions. To overcome the inherent limitations of polymer-based biomaterials and small molecule-based drug delivery systems, my postdoctoral work has focused on developing a recombinant self-assembled protein-based nanocarrier that can deliver sufficient amounts of functional antibodies to the cytosol for intracellular targeted labelling and manipulation of disease related proteins in live cells. I will use my unique microbiology-cell biology-material engineering cross-disciplinary approach to understand how bacteria and cells interact with biomaterials and exploit this insight for therapeutic purposes in tissue regeneration, bacterial infection and cancer therapy. My lab will also work at the intersection of drug delivery and materials engineering in order to develop novel bioresponsive/bioactive biomaterials and drug delivery systems. Specifically, my research will focus on three major areas. 1) Understanding the interactions between local microbes and host cells in a biomaterial environment is still in its infancy. I will elucidate the fundamental mechanisms of these interactions and apply this knowledge in design of advanced biomaterials/drug delivery systems for orthopedic infections and cancer therapy development. 2) I will apply modular design strategies to engineered protein biomaterials, which can be easily modified with multiple peptide domains containing different functionalities to mimic the diverse properties of the natural extracellular matrix. 3) I will use recombinant protein technologies to address a key bottleneck in biomolecule delivery, intracellular delivery of biomolecules such as therapeutic proteins and nucleic acids.

Education: Ph.D. in Biomedical Engineering. University of South Dakota, Vermillion, SD. (2012-2015) M.S in Biomedical Engineering. University of South Dakota, Vermillion, SD. (2010-2012)

Research/Work Experience: 2017-present, Postdoctoral Fellow, Georgia Institute of Technology, Atlanta, GA 2015-2017, Postdoctoral Fellow, University of South Dakota, Vermillion, SD

Selected Publications: • Wei Lv, Julie Champion. A modular platform for intracellular targeted labeling and antigen manipulation (In preparation) • Bo Yang, Wei Lv and Ying Deng. Drug loaded poly(glycerol sebacate) as a local drug delivery system for the treatment of periodontal disease, RSC Advance., 2017, 37426-37435 • Wei Lv, Jie Luo, Ying Deng, Yuyu Sun. Biomaterials immobilized with chitosan for rechargeable antimicrobial drug delivery. Journal of Biomedical Materials Research Part A. 2013, 447-455 • Jing Bao*, Wei Lv*, YuYu Sun, Ying Deng. 2013. Electrospun antimicrobial microfibrous scaffold for annulus fibrosus tissue engineering. Journal of Materials Science. 2013, 4223-4232. (* Equal Contribution) • Jie Luo, Wei Lv, Ying Deng, Yuyu Sun. Cellulose-ethylenediaminetetraacetic acid conjugates protect mammalian cells from bacterial cells. Biomacromolecules. 2013, 1054-1062

Awards/Honors: University of South Dakota Research & Creative Activity Grant (2014) Society for Biomaterials University of South Dakota Biomaterials Day Grant (2015) JOHNATHAN LYON, PhD BME, Duke University, 101 Science Drive, Durham, NC, 27705 . [email protected]

Research Overview: Dr. Lyon's research is currently focused on mechanisms and therapeutic systems resulting from the use of electrical regimes to control cellular behaviors and phenotypes. Dr. Lyon's thesis work culminated in a study of tumor cell migration under applied electrical fields (a phenomenon known as electrotaxis). Dr. Lyon pioneered assays for long-term, population density electrotactic analysis of cancer spheroids, which led to further characterization of underlying mechanisms in the differential responses in glioblastoma and medulloblastoma cells (through consequent RNA-seq and pharmaceutical neutralization/inhibition assays). Dr. Lyon has also contributed to several other courses of scientific inquiry, collaborating on projects involving biomaterial-based immunomodulation for peripheral nerve repair and the control of cancer growth, bacterial and viral carriers for brain cancer drug and gene delivery, bfrain-computer interfaces for robotic control, and he is also engaged in scientific outreach and teaching through his previous service and mentorship work, and via his recent appointment to the Duke Institute for Science & Society. Dr. Lyon is currently interested in expanding his work on electrical stimulation to develop generalizable bioelectronic systems to manipulate other cancer cells types–with the goal of producing effects beyond just migratory control–as well as to modulate microglial and macrophage phenotypes in disease- or injury- associated inflammation. His current work involves development of an in vivo electrosynthesizer to produce a programmable chorus of electrotherapeutic signals for targeted tissues in the brain, as well as a deeper study of how electrical stimulation montages can be shaped to better direct cells into or out of tissues.

Education: 2017 PhD, Biomedical Engineering, Georgia Tech/Emory University 2010 MS, Computer Science, University of Washington 2009 BS, Computer Science, University of Washington 2006 BFA, Digital and Experimental Arts and Media, University of Washington

Research/Work Experience: Jul 2019-Present Associate/Instructor, Institute for Science & Society, Duke University Oct 2017-Present Research Scientist, BME, Duke University Sep 2010-Oct 2017 Graduate Research Asst., Neuro. Biomat. and Cancer Therapeutics Lab (PI: Ravi Bellamkonda), Georgia Tech Jul 2015-Mar 2016 Graduate Intern/External Consultant, Takeda Pharmaceuticals May 2009-Jun 2010 Graduate Research Assistant, Neural Systems Laboratory (PI: Rajesh Rao), University of Washington May 2007-Jul 2009 Senior Computing Specials/Technical Coordinator, DXARTS, University of Washington

Selected Publications: Lyon, J. G., Carroll, S. L., Mokarram, N., & Bellamkonda, R. V. (2019). Electrotaxis of Glioblastoma and Medulloblastoma spheroidal Aggregates. Scientific reports, 9(1), 5309. Saxena, T., Lyon, J. G., Pai, S. B., Pare, D., Amero, J., Karumbaiah, L., ... & Bellamkonda, R. V. (2019). Engineering Controlled Peritumoral Inflammation to Constrain Brain Tumor Growth. Advanced healthcare materials, 8(4), 1801076. Mokarram, N., Dymanus, K., Srinivasan, A., Lyon, J. G., Tipton, J., Chu, J., ... & Bellamkonda, R. V. (2017). Immunoengineering nerve repair. Proceedings of the National Academy of Sciences, 114(26), E5077-E5084. Lyon, J. G., Mokarram, N., Saxena, T., Carroll, S. L., & Bellamkonda, R. V. (2017). Engineering challenges for brain tumor immunotherapy. Advanced drug delivery reviews, 114, 19-32. Mehta, N., Lyon, J. G., Patil, K., Mokarram, N., Kim, C., & Bellamkonda, R. V. (2017). Bacterial carriers for glioblastoma therapy. Molecular Therapy-Oncolytics, 4, 1-17.

Awards/Honors: 2011-2015 Norman & Rosalyn Wells Fellowship for Brain Tumor Research 2015 Executive Vice President of Research GTRIC Award 2012-2013 Georgia Tech Graduate Leadership Program Fellow 2011-2013 NIH Training Program Fellow in Cellular and Tissue Engineering 2011 NSF GRF Honorable Mention LEYUAN MA, PhD Koch Institute, MIT, 35 Adams St, Brookline, MA, 02446. [email protected]

Research Overview: Malfunction of the immune system leads to many diseases, such as cancer, autoimmunity, and severe infections. An exciting approach for addressing these issues is modulating the immune system using engineered biomolecules and immune cells. The vision for my future research program is to employ these modalities to further understand the immune system involvement in human diseases, with the ultimate goal of transforming biomolecules and immune cells into powerful immunotherapeutics.

Education: 09/2004 – 07/2008 B.S. Biology. Shandong Normal University, China 09/2009 – 12/2016 Ph.D. Biomedical Sciences, HHMI, UMass Medical School, USA. 12/2016 – present Postdoctoral Fellow, HHMI, Massachusetts Institute of Technology, USA.

Research/Work Experience: 11/2006 – 07/2008 Laboratory of Chengqiang He, Shandong Normal University • Characterization of the adaptive evolution of animal viruses under the selection pressure from host and vaccine. 10/2008 – 07/2009 Laboratory of Ligang Wu, Chinese Academy of Sciences • Mechanism of miRNA mediated deadenylation and translational repression in mammalian cells. 09/2009 – 02/2010 Laboratory of Phillip D. Zamore, Ph.D. HHMI, UMass Medical School • Identification of 3’-5’ exoribonucleases and terminal uridylyl transferases involved in miRNA degradation and tailing. 03/2010 – 12/2016 Laboratory of Michael R. Green, M.D. Ph.D. HHMI, UMass Medical School • Dissecting the mechanisms of drug resistance in leukemia using genome-wide shRNA screen, single-cell RNA-seq, and CRISPR-Cas9-based saturated mutagenesis. 12/2016 – present Laboratory of Darrell J. Irvine, Ph.D. HHMI, Massachusetts Institute of Technology • Bioengineering and Immunotherapy

Selected Publications: 1. L. Ma, T. Dichwalkar, J. Y. H. Chang, B. Cossette, D. Garafola, A. Q. Zhang, M. Fichter, C. Wang, S. Liang, M. Silva, S. Kumari, N. K. Mehta, W. Abraham, N. Thai, N. Li, K. D. Wittrup, D. J. Irvine, Enhanced CAR–T cell activity against solid tumors by vaccine boosting through the chimeric receptor, Science 365, 162–168 (2019). 2. N. Momin, N. K. Mehta *, N. R. Bennett *, L. Ma *, J. R. Palmeri, M. M. Chinn, E. A. Lutz, B. Kang, D. J. Irvine, S. Spranger, K. D. Wittrup, Anchoring of intratumorally administered cytokines to collagen safely potentiates systemic cancer immunotherapy, Sci Transl Med 11, eaaw2614 (2019). [* Equal contribution] 3. L. Ma*, M. L. Pak*, J. Ou, J. Yu, P. St Louis, Y. Shan, L. Hutchinson, S. Li, M. A. Brehm, L. J. Zhu, M. R. Green, Prosurvival kinase PIM2 is a therapeutic target for eradication of chronic myeloid leukemia stem cells, Proc. Natl. Acad. Sci. U.S.A. 116, 10482–10487 (2019). [* Equal contribution] 4. L. Ma, J. I. Boucher, J. Paulsen, S. Matuszewski, C. A. Eide, J. Ou, G. Eickelberg, R. D. Press, L. J. Zhu, B. J. Druker, S. Branford, S. A. Wolfe, J. D. Jensen, C. A. Schiffer, M. R. Green, D. N. Bolon, CRISPR-Cas9-mediated saturated mutagenesis screen predicts clinical drug resistance with improved accuracy, Proc. Natl. Acad. Sci. U.S.A. 114, 11751– 11756 (2017). 5. L. Ma, J. Roderick, M. A. Kelliher, M. R. Green, High-Throughput Screening of Tyrosine Kinase Inhibitor Resistant Genes in CML, Methods Mol. Biol. 1465, 159–173 (2016). 6. L. Ma, Y. Shan, R. Bai, L. Xue, C. A. Eide, J. Ou, L. J. Zhu, L. Hutchinson, J. Cerny, H. J. Khoury, Z. Sheng, B. J. Druker, S. Li, M. R. Green, A therapeutically targetable mechanism of BCR-ABL-independent imatinib resistance in chronic myeloid leukemia, Sci Transl Med 6, 252ra121–252ra121 (2014). 7. T. R. Wagenaar *, L. Ma *, B. Roscoe, S. M. Park, D. N. Bolon, M. R. Green, Resistance to vemurafenib resulting from a novel in the BRAFV600E kinase domain, Pigment Cell Melanoma Res 27, 124–133 (2014). [* Equal contribution]

Awards/Honors: 2014 Sigma Xi Grant-in-Aid of Research 2015 Dean’s Award for most insightful mid-thesis research 2016 Chinese Government Award for Outstanding Self-Financed Students Abroad 2019 American Cancer Society Postdoctoral Fellowship Award CHRISTOPHER M. MADL, PhD Baxter Laboratory for Stem Cell Biology, Stanford University, 269 Campus Drive, CCSR 4215, Stanford, CA, 94305 [email protected] Research Overview: Dynamic maintenance of the extracellular matrix (ECM) is crucial to proper tissue homeostasis. When the healthy wound healing response goes awry, aberrant ECM deposition leads to fibrosis, causing reduced tissue functionality and, in some cases, organ failure. Despite the critical role of ECM remodeling in regeneration and disease, present in vitro models are unable to adequately capture the dynamic and complex changes in ECM mechanics and biochemistry that occur in vivo. I apply a multifaceted strategy, drawing on protein engineering, bioorthogonal chemistries, and stem cell biology, to elucidate unappreciated mechanisms behind cell-ECM interactions impacting homeostasis and disease. During my PhD, I leveraged the well-defined properties of recombinant elastin-like proteins to selectively tune matrix stiffness and degradability independent of hydrogel microstructure, swelling, nutrient transport, and cell adhesive ligand concentration. This led to the unexpected observation that matrix degradation and remodeling are required for neural stem cells to maintain their stem cell phenotype and to subsequently differentiate into mature, functional neurons. I also developed site-selective techniques for introducing bioorthogonal reaction partners into engineered protein materials to enable gentle encapsulation of numerous cell types, while retaining the ability to independently tune matrix properties. As a postdoctoral fellow, I am characterizing the interplay between extrinsic mechanical changes in the stem cell niche and intrinsic defects in aged muscle stem cells. As part of this project, I developed a novel hydrogel system that enables real-time tracking of individual cells as matrix stiffness is altered by user- selected, minimally invasive stimuli. My independent research will develop and utilize dynamic hydrogel biomaterials to better understand how cells respond to fibrotic changes in their mechanical and biochemical microenvironments, using patient-derived organotypic cultures to identify therapeutic interventions for monogenic fibrotic diseases, such as pulmonary fibrosis.

Education: • Stanford University, September 2017. PhD, Bioengineering • Harvard University, May 2012. SM, Engineering Sciences • Harvard University, May 2012. AB, Engineering Sciences/Chemistry, summa cum laude

Research/Work Experience: • Stanford University School of Medicine, 2017 – Present Postdoctoral Fellow, Baxter Laboratory for Stem Cell Biology; Advisor: Helen M. Blau, PhD • Stanford University, 2012 – 2017 Graduate Research Assistant, Department of Bioengineering; Advisor: Sarah C. Heilshorn, PhD • Harvard University, 2008 – 2012 Undergraduate Research Assistant, School of Engineering & Applied Sciences; Advisor: David J. Mooney, PhD

Selected Publications: 1. Madl CM, Heilshorn SC, Blau HM. Bioengineering strategies to accelerate stem cell therapeutics. Nature. 557:335 (2018). 2. Madl CM, LeSavage BL, Dewi RE, Dinh CB, Stowers RS, Khariton M, Lampe KJ, Nguyen D, Chaudhuri O, Enejder A, Heilshorn SC. Maintenance of neural stemness in 3D hydrogels requires matrix remodeling. Nature Materials. 16:1233 (2017). 3. Madl CM, LeSavage BL, Dewi RE, Lampe KJ, Heilshorn SC. Matrix remodeling enhances the differentiation capacity of neural progenitor cells in 3D hydrogels. Advanced Science. 6:1801716 (2019). 4. Madl CM, Katz LM, Heilshorn SC. Bio-orthogonally crosslinked, engineered protein hydrogels with tunable mechanics and biochemistry for cell encapsulation. Advanced Functional Materials. 26:3612 (2016). 5. Madl CM, Mehta M, Duda GN, Heilshorn SC, Mooney DJ. Presentation of BMP-2 mimicking peptides in 3D hydrogels directs cell fate commitment in osteoblasts and mesenchymal stem cells. Biomacromolecules. 15:445 (2014).

Awards/Honors: • Life Sciences Research Foundation (LSRF) Postdoctoral Fellowship. 2019 – 2022. • National Research Service Award Postdoctoral Fellowship (F32). NIH/NIA. 2019 – 2022. (Declined to accept LSRF) • Early Career Editorial Advisory Board, ACS Biomaterials Science & Engineering. 2018 – 2021. • Stanford ChEM-H Postdoctoral Training Program in Quantitative Mechanobiology. 2017 – 2019. • Siebel Scholar, Class of 2017. • National Research Service Award Predoctoral Fellowship (F31). NIH/NIBIB. 2015 – 2017. • Thomas Hoopes Senior Thesis Prize and Phi Beta Kappa, Harvard University, 2012. BHUSHAN MAHADIK, PhD Department of Bioengineering, University of Maryland, College Park, 8278 Paint Branch Dr., College Park, MD 20742 [email protected]

Research Overview: The fields of tissue engineering and regenerative medicine hold great promise for transforming human lives. As a chemical engineer with a background in materials science and bioengineering my goal is to improve our understanding of the intricacies of native biology while also providing clinically translatable solutions. My graduate research aimed at engineering the hematopoietic stem cell (HSC) niche within the bone marrow. These cellsplay an integral role in reconstituting all the blood and immune cells and are an important component of bone marrow transplants. Using complementary strategies such as microfluidics, tethered biomolecules, and ECM proteins, we examined the role of small molecule signaling and bone marrow niche environment on HSC lineage specification and differentiation. During my postdoctoral work, I also developed mathematical models to better elucidate the feedback mechanisms driving this process. At my current position as the Assistant Director for the NIH Center for Engineering Complex Tissues, I am employing biomaterial, 3D printing, and bioreactor strategies to develop in vitro models for investigating bone, liver, and vascular physiologies in a highly collaborative environment. My experience has taught me that collaboration between engineers, scientists, and clinicians can lead to truly transformative research. The long-term research goal and focus of my laboratory will be on investigating the role of 1) native microenvironment 2) physiological stimuli and 3) tissue form and function on cellular response. We will leverage tissue engineering and 3D printing strategies to examine the native bone marrow HSC microenvironment, its dysregulation leading to various cancers and disorders, and the role of vascularization in various pathologies. The overarching aim of our research will be to develop biologically relevant in vitro models that help elucidate underlying mechanisms driving cellular and organ- level processes and strategies for clinical translation of these tissues.

Education: Ph.D. Chemical Engineering, University of Illinois at Urbana-Champaign; Advisor: Dr. Brendan Harley 2008 – 2014 BS, Chemical Engineering and Materials Science Engineering, University of California, Berkeley 2004 – 2008

Research/Work Experience: Assistant Director, NIH Center for Engineering Complex Tissues, University of Maryland 2017 – present Postdoctoral Research Associate, Dept. of Chemical Engineering, Univ. of Illinois at Urbana Champaign 2014 – 2017 Graduate Research Assistant, Dept. of Chemical Engineering, Univ. of Illinois at Urbana Champaign 2008 – 2014 Undergraduate Research Assistant, lab of Dr. Clayton Radke, University of California, Berkeley 2007 – 2008 Undergraduate Research Assistant, lab of Dr. Alexander Katz, University of California, Berkeley 2006 – 2007

Selected Publications: Mahadik B, Hannon B, Harley BAC, “A computational model of feedback-mediated hematopoietic stem cell differentiation in vitro”, PLoS One, 2019 Mar 1;14(3) Mahadik B, Bharadwaj NA, Ewoldt RH., Harley BAC, “Regulating dynamic signaling between hematopoietic stem cells and niche cells via a hydrogel matrix” Biomaterials, 2017;125; 54-64 Choi JS (co), Mahadik B (co), and Harley BAC, “Engineering the hematopoietic stem cell niche: frontiers in biomaterial science,”Biotechnology Journal, 2015;10;1529-45, Mahadik B, Pedron S, Skertich L, and Harley BAC, “The use of covalently immobilized stem cell factor to effect hematopoietic stem cell activity within a 3D gelatin hydrogel,” Biomaterials, 2015;67; 297-307 Mahadik B, Wheeler T, Skertich L, Kenis PJA, and Harley BAC, “Microfluidic generation of gradient hydrogels to modulate hematopoietic stem cell culture environment,” Advanced Healthcare Materials, 2014;3:449-58

Honors and Awards: Outstanding Abstract Award, Orthopaedic Biomaterials SIG, Society For Biomaterials 2019 Annual Meeting, Seattle, WA 2019 1st Place, Postdoctoral Presentation Competition, MRL Biological Conference, UIUC 2016 $50,000 Proof-of-Concept Award and Illinois NSF I-Corps selection 2015 1st Place, Student & Young Investigator Poster Competition, TERMIS World Congress 2015 Hanratty Travel Award, Chemical Engineering Department, UIUC 2010, 2013 Melvin J. Heger-Horst Scholarship, College of Chemistry, UC Berkeley 2006 Asteroid renamed to Minor Planet (17095) Mahadik, MIT Lincoln Laboratory, ISEF 2005 KALPANA MANDAL, PhD Institute for Medicine and Engineering, University of Pennsylvania, 1080 Vagelos Research Building, Philadelphia, PA, 19104 [email protected]

Research Overview: My current research focuses on understanding cellular mechanoresponses when cells are in a disease state by using a variety of 2D and 3D substrates including elastic or viscoelastic PAA hydrogels, hyaluronic acid gels, collagen gels, Gelatin and fibrin gels. Understanding the rheological properties of biopolymer network and their contributions to tissue mechanics is my interest. In addition, my research showed how intracellular mechanical properties are altered during disease development by using active microrheology with optical tweezers. Moreover, the intracellular reorganization and cellular traction forces on the extracellular matrix in response to geometrical cues has be shown by combining traction force microscopy and substrate micropatterning.

Education: 2015/10-present Postdoc, University of Pennsylvania 2012/10 – 2015/06 Postdoc, Institute Curie Paris 2009/09 – 2012/09 Ph.D. Biophysics, Grenoble University, Grenoble, France. 2007 M.Sc. Physics (honors), Indian Institute of Technology Kanpur, India

Research/Work Experience: 2015-present: University of Pennsylvania, Philladelphia- (Postdoc) Advisor: Prof. Paul A Janmey Subject: Cancer cell mechanosenseing and measuring the cell mechanical properties 2012-2015: Institute Curie, Paris, France – (Postdoc) Advisor: Prof. Bruno Goud and Prof. Jean-Baptiste Manneville Subject: Probing intracellular micro-rheological properties using optical tweezers. 2009-2012: Laboretoire Interdesciplinarie de Physique, CNRS,Grenoble,France - Physics for life Sciences (Ph.D.) Advisor: Dr. Martial Balland Subject: Role of ECM in cell internal organization using Traction Force Microscopy and micropatterning

Selected Publications: (‡ Corresponding authors) •Mandal K.‡, Pogoda K., Nandi S., Mathieu S., Kasri A., Radvanyi F., Goud B., Janmey P. A.‡, Manneville J.B.‡. Role of the KIF20A kinesin in the mechanics and migration of bladder cancer cells . Nano Letters 2019 (minor review) •Mandal K ‡, Aroush D.R, Graber Z., Wu B., Park CY., Fredberg J.J., Guo W., Baumgart T., Janmey P.A. ‡ . Soft hyaluronic gel promotes cell spreading, stress fibers, focal adhesion, membrane tension by phosphoinositide signaling, not traction force. ACS Nano 2018 •Mandal, K.; Asnacios, A.; Goud, B.; Manneville, J.-B. Mapping Intracellular Mechanics on Micropatterned Substrates. Proc. Natl. Acad. Sci. 2016, 113 (46), E7159–E7168. - Highlighted in Institute Curie news [curie news] •Mandal, K.; Wang, I.; Vitiello, E.; Orellana, L. A. C.; Balland, M. Cell Dipole Behaviour Revealed by ECM Sub- Cellular Geometry. Nat. Commun. 2014, 5, 5749. -Highlighted in leading French newspaper Le Monde [Press release] •Mandal, K*.; Guet, D*.; Pinot, M.; Hoffmann, J.; Abidine, Y.; Sigaut, W.; Bardin, S.; Schauer, K.; Goud, B.; Manneville, J. B. Mechanical Role of Actin Dynamics in the Rheology of the Golgi Complex and in Golgi-Associated Trafficking Events. Curr. Biol. 2014, 24 (15), 1700–1711. (*co-first author).-Commentary by Gustavo Egea, Carla Serra peinado Golgi Apparatus: Finally Mechanics Comes to Play in the Secretory Pathway. [ Commentary ], - Highlighted in the Institute [Curie news]

Awards/Honors: •2017/2019-present Reviewer, Soft Matter, Biophysical Journal, Journal of Materials Chemistry B •2018 BPP postdoctoral travel award by University of Pennsylvania •2016/2017 Abstract Reviewer, Poster judge, Biomedical postdoctoral research symposium, UPenn, USA •2016/2017 Invited/ poster judge, ASCB meeting, USA •2016 Travel award by ASCB.The company of Biologists, USA •2014-2015 Labex CelTisPhyBio Postdoctoral fellowship, France •2009-2012 RTRA Nanosciences foundation Doctoral fellowship, France

ALEXANDER E. MARRAS, PhD Pritzker School for Molecular Engineering, University of Chicago, 5640 S Ellis Ave, ERC 108, Chicago, IL, 60637 [email protected]

Research Overview: Biomolecular machinery communicates with physical and chemical signals to perform essential tasks in our cells. As engineers, we strive to mimic such functionality in an effort to build engineering devices and better understand nature. My graduate research expanded structural DNA nanotechnology by creating nanodevices with programmable motion. Borrowing concepts from macro-scale machine design, I developed nanoscale kinematic joints including hinges and sliders and used these to build higher order mechanisms with specific 2D and 3D motion. I then developed multiple actuation methods enabling real-time tunable control of DNA-based devices. My postdoctoral research uses charged homo- and block copolymers to bind nucleic acids or proteins forming nanoparticles and micro-scale materials. A thorough investigation into chemical structure-physical property and stability relationships of these complexes enables efficient design of therapeutic nanoparticles and programmable materials. Building on my cumulative research expertise, my future research plans will employ the programmability of DNA assembly and the tunability and robustness of polymer assemblies to investigate the mechanical and chemical interactions between biomolecules and their environment, employ nanoparticles for therapeutic biomolecule delivery, and develop predictable methods for multiscale assembly of nanostructures.

Education: PhD: Mechanical Engineering, Ohio State University – Aug 2017 MS: Mechanical Engineering, Ohio State University – May 2013 BS: Mechanical Engineering, Ohio State University – June 2011

Research/Work Experience: Postdoc: “Polyelectrolyte Complexation with Biomolecules” Advisor: Matt Tirrell, Molecular Engineering, University of Chicago Ph.D.: “Design, Control, and Implementation DNA Origami Mechanisms” Advisor: Carlos Castro, Mechanical Engineering, Ohio State University B.S.: “Finite Element Modeling of Edge Crack Growth” Advisor: Prasad Mokashi, Mechanical Engineering, Ohio State University Industry Internships: Finite Element Modeling at Honda R&D; Product Design at Bose

Selected Publications: (Chronological, of 14 total journal articles, Google Scholar h-index: 10): 1. Marras, A.E., Vieregg, J.R., Ting, J.M., Rubien, J.D., Tirrell, M.V. “Polyelectrolyte complexation of oligonucleotides by charged hydrophobic – neutral hydrophilic block copolymers” Polymers. 11:83 (2019) 2. Marras, A.E., Shi, Z., Lindell, M., Patton, R.A., Huang, C.M., Zhou, L., Su, H-J., Arya, G., Castro, C.E. “Cation- activated avidity for rapid reconfiguration of DNA nanodevices” ACS Nano. 12:9484-9494 (2018) 3. Lei, D., Marras, A.E., Liu, J., Huang, C.M., Zhou, L., Castro, C.E., Su, H.J., Ren, G. “Three-dimensional structural dynamics of DNA origami Bennett linkages using individual-particle electron tomography” Nature Communications. 9:592 (2018) 4. Marras, A.E., Zhou, L., Kolliopoulos, V., Su, H.J., Castro, C.E. “Directing folding pathways for multi-component DNA origami nanostructures with complex topology.” New Journal of Physics. 18:055005 (2016) 5. Marras, A.E., Zhou, L., Su, H.J., Castro, C.E. “Programmable motion of DNA origami mechanisms.” Proceedings of the National Academy of Sciences. 112:713-8 (2015) a. Highlighted in Nature Materials, Nature Nanotechnology, PNAS

Awards/Honors: • Presidential Fellowship, Ohio State University - 2017 • Best Student Poster Award, OSU Institute for Materials Research - 2017 • Ray Travel Award, Ohio State University - 2017 • Education Travel Award, Biophysical Society - 2017 • Ray Travel Award Ohio State University - 2015 • Best Poster Award, Foundations of Nanoscience (FNANO) – 2014 • Graduate Fellowship, Ohio State University - 2012-2014 JOHN MARTIN, PhD Orthopaedic Surgery, Duke University, Medical Sciences Research Building I, DUMC Box 3093, Durham, North Carolina, 27710. [email protected] Research Overview: I am a mechanical engineer by training with interests in spine function and low back pain. Overall, my skillset includes human subjects research, animal models, biomechanics, tissue engineering, and in vivo imaging. As a graduate student, I developed expertise in soft tissue mechanics, specifically of the intervertebral disc, and in animal models for disc degeneration, regeneration, and tissue engineering. I am currently a postdoctoral researcher at Duke University, where I am developing magnetic resonance imaging techniques to measure intervertebral disc geometry, composition, and function as well as high-speed radiography techniques to measure in vivo disc function in humans. My goal is to develop a lab with two focuses. The first will be a translational pipeline in which the lab develops and tests strategies to improve spine health in animal models and seamlessly evaluates those in humans. The second will be to identify the underlying causes of spine disease in humans.

Education: (1) BS Mechanical Engineering 2006 - The College of New Jersey (2) MS Mechanical Engineering 2008 - University of Colorado (3) PhD Mechanical Engineering and 2015 - University of Pennsylvania

Research/Work Experience: (1) 2006 - 2008, Graduate Student, University of Colorado Advisor: Virginia Ferguson; Research area: Blood vessel mechanical function (2) 2008 - 2009, Research Engineer, University of Pennsylvania Advisor: Dawn Elliott, PhD; Research area: Intervertebral disc mechanical function (3) 2010 - 2015, Graduate Student, University of Pennsylvania Advisor: Rob Mauck, PhD; Research area: Intervertebral disc tissue engineering (4) 2016 - present, Ruth L. Kirschtein NRSA Postdoctoral Fellow, Duke University Advisor: Lou DeFrate, ScD; Research area: In vivo musculoskeletal imaging and mechanical function

Selected Publications: (1) Martin JT, DeFrate LE, et al. (2018) A magnetic resonance imaging framework for quantifying intervertebral disc deformation in vivo: reliability and application to diurnal variations in lumbar disc shape. J Biomech, 71:291-295 (2) Martin JT, Mauck RL, et al. (2017) In vitro maturation and in vivo integration and function of an engineered cell- seeded disc-like angle Ply structure (DAPS) for total disc arthroplasty. Sci Rep 7(1):15765 (3) Martin JT, Elliott DM et al. (2013) Needle puncture injury causes acute and long-term mechanical deficiency in a mouse model of ntervertebral disc degeneration. J Orthop Res; 31:1276–1282

Awards/Honors: 2019 Best Poster Award, Orthopaedic Research Society Annual Meeting (Spine Section) 2018 Best Poster Award, Philadelphia Spine Research Symposium 2018 New Investigator Recognition Award, Orthopaedic Research Society Annual Meeting 2017 New Investigator Recognition Award (Co-Author), Orthopaedic Research Society Annual Meeting 2017 Best Poster Award, Orthopaedic Research Society 4th International Spine Research Symposium 2015 Poster Competition 2nd Place, Philadelphia Spine Research Symposium 2015 “Art in Science” Competition Co-Winner, Perelman School of Medicine, University of Pennsylvania 2015 PhD Paper Competition 2nd Place, Summer Biomechanics, Bioengineering and Biotransport Conference 2014 Best Presentation Award, Philadelphia Spine Research Symposium 2013 Spotlight Presentation (Co-author), Orthopaedic Research Society Annual Meeting 2012 Force and Motion Academic Scholarship ($10,000) 2012 PhD Paper Competition Finalist, American Society of Mechanical Engineers, Summer Bioengineering Conference 2012 Conference Travel Grant, Graduate and Professional Student Assembly, University of Pennsylvania 2012 Spotlight Presentation, Orthopaedic Research Society Annual Meeting 2010 Best Poster Award, Orthopaedic Research Day, University of Pennsylvania MATTHEW D.J. McGARRY, PhD Dartmouth College, 63 East Wheelock St, Hanover, NH, 03755 . [email protected]

Research Overview: The majority of disease processes alter the structure of tissue, which in turn alters the mechanical properties with potentially very large contrasts between healthy and diseased tissue. Measuring these mechanical properties can provide new, highly sensitive diagnostic indicators. The overarching theme of my research has been fitting mechanical models to large imaging datasets to extract relevant tissue parameters. In my career to date, I have investigated a wide range of models, tissue types, and sources of data for many applications particularly MRI and Ultrasound in the brain and cardiovascular system. My lab will continue this path through three main research phases. The first phase of my research will apply my successful approach to MR elastography developed together with my collaborators to open clinical problems. Advantages of my Nonlinear Inversion (NLI) method over competing approaches include accurate treatment of heterogeneity, adaptability to a range of mechnical models, and a rich library of possible reguolarization and stabilization techniques. We have had substantial success in the brain, where we have found strong correlations between the damping ratio of brain structures such as the hippocampus, orbitofrontal cortex, and ventromedial prefrontal cortex with performance in associated cognitive tests (lower-functioning brains are more viscous). This finding has been replicated in multiple studies at two different sites. The unprecedented ability to image a structural correlate of brain function in vivo has an enormous range of clinical applications, including Alzheimer’s, aging, multiple sclerosis, and brain development, which I am currently pursuing with several collaborators. My model-based imaging approach is amenable to a range of imaging modalities, including MRI, ultrasound, optical coherence tomography (OCT), and confocal microscopy. In the second phase I will develop multi-modality elastography to examine the same diseased tissues across multiple scales – for example, is the observed decreased arterial compliance in hypertension primarily driven by the intima, media or adventitia layers of the artery? Is the decreased macroscale stiffness observed in degenerative brain diseases from changes in the cell membranes, cell bodies, neurons or extracellular matrix? This information could help develop and monitor targeted therapies. The higher risk, higher reward part of my research plan is development of “virtual histology” by fitting microscale parameters of multi-scale forward models to in vivo displacement measurements from conventional imaging modalities. This will generate maps of parameters that directly reflect tissue organization in living subjects rather than the more abstract continuum mechanical properties such as shear and loss modulus in conventional elastography.

Education: Ph.D., Dartmouth College 2013 M.E., University of Canterbury, New Zealand, 2008 B.E. (Hons), , University of Canterbury, New Zealand, 2008 (Major: Mechanical Engineering)

Research/Work Experience: Areas of Expertise: Numerical methods in engineering, including programming finite element, boundary element and finite difference methods, and integrating these methods with optimization techniques to fit multiparametric models to experimental data. MR Elastography: 2008-2019, Dartmouth College. Supervisor: Keith Paulsen. University of Canterbury:2006-2008. Supervisor: Eli Van Houten Ultrasound Elasticity Imaging: Columbia University, 2015-2017. Supervisor: Elisa Konofagou.

Selected Publications: McGarry, M. D. J., et al. "Multiresolution MR elastography using nonlinear inversion." Medical physics 39.10 (2012): 6388-6396. McGarry, M. D. J., et al. "Suitability of poroelastic and viscoelastic mechanical models for high and low frequency MR elastography." Medical physics 42.2 (2015): 947-957. McGarry, Matthew, et al. "Including spatial information in nonlinear inversion MR elastography using soft prior regularization." IEEE transactions on medical imaging 32.10 (2013): 1901-1909. McGarry, Matthew, et al. "Uniqueness of poroelastic and viscoelastic nonlinear inversion MR elastography at low frequencies." Physics in Medicine & Biology 64.7 (2019): 075006. Mcgarry, Matthew, et al. "An inverse approach to determining spatially varying arterial compliance using ultrasound imaging." Physics in Medicine & Biology 61.15 (2016): 5486. Johnson, Curtis L.,McGarry, Matthew et al. "Double dissociation of structure-function relationships in memory and fluid intelligence observed with magnetic resonance elastography." Neuroimage 171 (2018): 99-106.

Awards/Honors: Wrote Dartmouth's portion of two funded NIH R01 grants. Patent: Poroelastic dynamic mechanical analyzer for determining mechanical properties of biological materials. Thayer School of Engineering at Dartmouth - Alma Hass Milham Fellowship, 2010-2011 - Graduate Research Fellowship, 2008-2010 University of Canterbury (New Zealand) - C.W. Hamilton Masters Scholarship 2006 JOUHA MIN, PhD Center for Systems Biology, MGH/Harvard Medical School, Boston, MA 02114 [email protected]

Research Interests: Precision or personalized medicine is an emerging approach that matches patients with the most appropriate treatment based on the precise molecular features of an individual patient’s disease. Despite huge advances in translational research, diagnosis and treatment continue to pose profound challenges in addressing significant health problems, including cancer and infectious diseases. In the MTFC presentation, I will showcase three research projects that demonstrate molecular engineering efforts toward precision medicine. Two focus on new developments in diagnosis: (i) bioanalytical sensing of plasma markers predictive of impending sepsis based on a magnetoelectrochemical approach, and (ii) molecular profiling of breast cancer assisted by deep-learning algorithms for global health and point-of-care diagnostics. The third utilizes a novel programmable multi-therapy release strategy for orthopedic implants, enabling both bacterial eradication at the bone-hardware interface and accelerated bone tissue regeneration. These studies highlight the potential of integrating materials science, molecular engineering, and clinical science approaches to develop advanced molecular diagnostics and innovative therapies for precision medicine.

Educations: MIT, 2016, PhD in Chemical Engineering Cornell University 2010, BS in Chemical Engineering

Research Experience: • MGH/Harvard Medical School, 2016-present NIH T32 Postdoctoral Research Fellow (Advisor: Ralph Weissleder, MD PhD) • MIT, 2010-2016 Graduate student (Advisors: Paula T. Hammond, PhD and Richard D. Braatz, PhD) • Cornell University, 2008-2010 Undergraduate research assistant (Advisors: Paulette Clancy, PhD / Jonathan Butcher, PhD)

Selected Publications: • Min J†, Im H†, Allen M, McFarland PJ, Degani I, Yu H, Normandin E, Pathania D, Patel JM, Castro CM, Weissleder R*, Lee H*, Computational optics enables breast cancer profiling in point-of-care settings, ACS Nano, 12(9), 9081-9090 (2018). †equal contribution • Min J, Nothing M, Coble B, Zheng H, Park J, Im H, Weber G, Castro CM, Swirski FK, Weissleder R*, Lee H*, Integrated biosensor for rapid and point-of-care sepsis diagnosis, ACS Nano, 12(4), 3378-3384 (2018). • Min J, Choi KY, Dreaden EC, Padera RF, Braatz RD, Spector M, Hammond PT, Designer dual therapy implant coatings eradicate biofilms and accelerate bone tissue repair, ACS Nano, 10(4), 4441-4450 (2016) Min J, Braatz RD, Hammond PT, Tunable staged release of therapeutics from layer-by-layer coating with clay interlayer barrier, Biomaterials, 35(8), 2507-17 (2014).

Awards/Honers: • Poster Award (1st Place, Travel Grant $1,000), MGH Scientific Advisory Committee (SAC) meeting, 2018 • MRS Poster Award Winner (1st place), Materials Research Society (MRS) Fall Meeting, 2015 • KSEA-KUSCO Graduate Scholarship, 2014 • Graduate Scholarship, Mokam Life Science Research, Green Cross Holding Corp., 2013 • Merrill Presidential Scholarship, Cornell University, 2010 JENNIFER A. MITCHEL, PhD Molecular and Integrative Physiology, Harvard University, 665 Huntington Ave, Boston, MA, 02115 [email protected]

Research Overview: My research focuses on the mechanics of collective epithelial migration, epithelial plasticity, and emergent mesoscale phenomena. Every organ surface and body cavity is lined by a confluent collective of epithelial cells. In homeostatic circumstances the epithelial collective remains effectively solid-like and sedentary. But during morphogenesis, remodeling or repair, as well as during malignant invasion or metastasis, the epithelial collective becomes fluid-like and migratory. The plasticity of epithelial cells which allows them to shape tissues, repair wounds, and invade during metastasis is commonly understood to be governed by the epithelial-tomesenchymal transition (EMT). EMT is an epigenetic program entailing progressive loss of epithelial character accompanied by progressive gain of mesenchymal character, whereby each epithelial cell tends to free itself from adhesions to immediate neighbors and acquires migratory capacity and invasiveness. However, my work has shown that in contrast to the widely accepted obligatory role for EMT in epithelial tissue plasticity and migration, differentiated epithelia are able to remodel –constituent cells migrate, change their neighbors, and exhibit emergent collective dynamics– without EMT. Instead, the cells utilize the newly discovered unjamming transition (UJT), in which cells retain epithelial integrity but change shape in order to migrate within the crowded collective. Our discovery that the cellular UJT is distinct from the EMT mechanism reveals a powerful, unifying, and overriding biological principle and may be the initial step towards reframing a core concept in epithelial cell biology. In my lab, we will bridge the gap between cellular biomechanics and collective epithelial migration through the lens of soft matter physics and the mechanism of cellular jamming, in the following projects: ·Project 1: Investigate the roles of adhesion, contractility, polarity and propulsion in epithelial solid-fluid transitions. ·Project 2: Determine the molecular and physical mechanisms of emergent collective migration during mechanically-induced UJT. ·Project 3: In vitro and in vivo, determine when UJT and EMT may act cooperatively, sequentially or independently to effect epithelial tissue sculpting.

Education: Ph.D. Biomedical Engineering, Brown University (2013) S.B. Mechanical Engineering, Massachusetts Institute of Technology (2007)

Research/Work Experience: Research Associate, Harvard University. 2016-Present. Mentor: Jeffrey Fredberg, Ph.D. NIH NHLBI Ruth L. Kirschstein T32 Postdoctoral Fellow, Harvard University. 2013-2016. Mentor: Jin-Ah Park, Ph.D. Graduate researcher, Biomedical Engineering, Brown University. 2007-2013. Mentor: Diane Hoffman-Kim, Ph.D. Undergraduate researcher, Biological Engineering, MIT. 2004-2007. Mentors: Drew Endy, Ph.D, and Linda Griffith, Ph.D. Selected Publications: [1] Mitchel JA, Das A, O’Sullivan MJ, Stancil IT, DeCamp SJ, Koehler S, Butler JP, Fredberg JJ, Nieto MA, Bi D, Park J-A. (2019) The unjamming transition is distinct from the epithelial-to-mesenchymal transition. BioRxiv. [2] Lan B*, Mitchel JA*, O’Sullivan MJ, Park CY, Kim JH, Cole WC, Butler JP, Park J-A. (2018) Airway epithelial compression promotes airway smooth muscle proliferation and contraction. AJP Lung Cellular and Molecular Physiology (*equal contribution). [3] Mitchel JA, Antoniak S, Lee JH, Kim SH, McGill M, Kasahara DI, Randell SH, Israel E, Shore SA, Mackman N, Park J-A. (2016) IL-13 Augments Compressive Stress-Induced Tissue Factor Expression in Human Airway Epithelial Cells. American Journal of Respiratory Cell and Molecular Biology. [4] Mitchel JA, Martin IS, Hoffman-Kim D. (2013) Neurient: an algorithm for automatic tracing of confluent neuronal images to determine alignment. Journal of Neuroscience Methods. [5] Mitchel JA, Hoffman-Kim D. (2011) Cellular Scale Anisotropic Topography Guides Schwann Cell Motility. PLoS ONE.

Awards/Honors: Parker B. Francis Fellowship, 2019 - 2022 American Thoracic Society Abstract Scholarship Award, 2017 Postdoc Association Travel Award, Harvard T.H. Chan School of Public Health, 2014 Robert and Susan Kaplan Fellowship, Brown University, 2011 GAANN Fellowship, Institute for Molecular and Nanoscale Innovation, Brown University, 2010 - 2012 Brown Institute for Brain Science Graduate Research Award, 2009 AARON H. MORRIS, PhD Biomedical Engineering, University of Michigan, 1600 Huron Pkwy, Ann Arbor, MI, 48109 [email protected]

Research Overview: The goal of my research program will be to harness innovative biomaterials-based approaches to study physiological and pathological processes, as well as create translational technologies. In my doctoral work, I focused on engineering material properties to control cell-material interactions and manipulate the host response. As a postdoctoral fellow, I leveraged this expertise to harness the host response to materials to create engineered tissue surrogates (ETS). I developed ETS that reflect the immune status during disease and enable early diagnosis and treatment monitoring in autoimmunity sufficient to intervene and prevent disease onset before symptoms occur. My laboratory will combine approaches learned in my doctoral and postdoctoral training in three ways: i) harnessing empty ETS to diagnose and monitopr diseases of aberrant immune function, ii) modifying ETS properties with drug encapsulation / materials modification strategies to create niches more similar to target organs and enrich pathological cell populations, and iii) developing and harnessing continuous monitors for immune status within the host that can be non-invasively monitored. Thus, my laboratory will harness biomaterials for translational as well as fundamental discoveries. My research program will be interdisciplinary – spanning biomaterials development, immunology, and regenerative medicine. My unique background in engineering material properties to tailor the foreign body response and harnessing the response to materials to interrogate the immune system have provided a niche in which my lab will impact patient care both by providing technologies for early detection and treatment of disease and by monitoring the response to therapy.

Education: Ph.D., Biomedical Engineering, 2017, Yale University M.S. and M.Phil, Biomedical Engineering, 2014 & 2015, Yale University B.S., Biomedical Engineering, 2012, Georgia Institute of Technology

Research/Work Experience: MLS Postdoctoral Fellow, Biomedical Engineering, University of Michigan 2017-Present Advisor: Lonnie Shea Graduate Research Assistant, Biomedical Engineering, Yale University, 2012-2017 Advisor: Themis Kyriakides Fellowship, Yale Entrepreneurial Institute, 2016

Selected Publications: Morris AH, Hughes KR, Oakes RS, Cai MM, Shea LD. Engineering tissue surrogates to monitor autoimmune progression, relapse, and treatment efficacy. Submitted. Morris AH, Stamer DK, Kunkemoeller B, Chang J, Xing H, Kyriakides TR. Decellularized materials derived from TSP2- KO mice promote enhanced neovascularization and integration in diabetic wounds. Biomaterials 169, (2018). Morris AH, Lee H, Xing H, Stamer DK, Tan M, Kyriakides TR. Tunable Hydrogels Derived from Genetically Engineered Extracellular Matrix Accelerate Diabetic Wound Healing. ACS Appl. Mater. Interfaces 10, 41892–41901 (2018). (Highlighted as ECM News Top Story) Morris AH, Mahal R, Udell J, Wu M, Kyriakides TR. Multi-compartment Drug Release System for Dynamic Modulation of Tissue Responses. Adv. Healthc. Mater. 6, (2017). Morris AH, Stamer DK, Kyriakides TR. The Host Response to Naturally-Derived Extracellular Matrix Biomaterials. Semin. Immunol. 1–20 (2017). Morris AH, Kyriakides TR. Matricellular proteins and biomaterials. Matrix Biol 2014.

Awards/Honors: Michigan Precision Health Scholars Grant 2018 Michigan Life Science Institute Fellowship 2018-Present NIH R.L. Kirschstein National Research Service Award Institutional Fellowship (T32) 2019 NSF Graduate Research Fellowship 2013 SBIR funded for startup from PhD work 2019 First Place Talk, UM Precision Health Symposium 2019 Second Place Poster (Most Transformative) UM Precision Health Symposium 2019 Yale Advanced Graduate Leadership Program Fellowship 2015-2017 Summer IRTA Fellowship, BESIP program, NIBIB, National Institutes of Health 2011 XUAN MU, PhD BME, Tufts University, 4 Colby St, Room 126, Medford, MA, 02155 . [email protected]

Research Overview: My research focuses on the interface between microfluidics, biomaterials, and biology for the modeling of physiological functions, as well as clinical diagnosis at the point of care. Recently, I am also interested in the aqueous solvent-directed assembly of silk protein that underpins 3D printing of neat proteinacious structures including microfluidic chips.

Education: Ph.D. in Applied Chemistry, 2010.6 East China University of Science and Technology, Shanghai, China B. S. in Applied Chemistry, 2005.9 East China University of Science and Technology, Shanghai, China

Research/Work Experience: Research Associate, 2017.1-present Department of Biomedical Engineering, Tufts University,Medford, MA Visiting Scholar and Research Fellow, 2016.2-2017.1 Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA Associate Professor, 2014.7-2016.2 Assistant Professor, 2012.6-2014.7 Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences School of Basic Medicine, Peking Union Medical College, Beijing, China Postdoctoral Fellow, 2010.6-2012.6 National Center for Nanoscience and technology, Beijing, China

Selected Publications: 1.Xuan Mu*, et al. A paper-based skin patch for the diagnostic screening of , Chemical Communications, 2015, 51, 6365-6368. Highlights in Chemical Communications Blog, American Association of Clinical Chemistry (invited blog) and BioNews Texas 2.Xuan Mu*, et al. Multiplex Microfluidic Paper-based Immunoassay for the Diagnosis of Hepatitis C Virus Infection, Analytical Chemistry, 2014, 86, 5338-5344. Highlights in American Chemical Society, The Analytical Scientist (invited editorial), ScienceDaily, phys.org, eurekAlert!, RedOrbit and Innovationtoronto 3.Xuan Mu, et al. Laminar Flow used as "Liquid Etching Mask" in Wet Chemical Etching to Generate Glass Microstructures with an Improved Aspect Ratio. Lab on a Chip, 2009, 9, 1994-1996. Highlights in Nature Materials 4.Xuan Mu, et al. Engineering 3D Vascular Network in Hydrogel for Mimicking a Nephron, Lab on a Chip, 2013, 13(8):1612-1618. 5.Xuan Mu, et al. Three-dimensional printing of silk protein structures by aqueous solvent-directed molecular assembly, Macromolecular Bioscience, 2019, accepted.

Awards/Honors: Peking Union Medical College Rising Star Award 2014 Distinguished Abstract Award of National Academy of Clinical Biochemistry 2013 International Travel Grant of VSF Foundation for AACC Annual Meeting 2013 Chinese Academy of Sciences K. C. Wong Post-doctoral Fellowship 2010 JENNA MUELLER, PhD Biomedical Engineering, Duke University, 101 Science Drive, Box 90281, Durham, NC, 27708, [email protected]

Research Overview: I am currently a Postdoctoral Associate in the Department of Biomedical Engineering at Duke University. My program of research centers on developing translational low-cost optical and therapeutic technologies to improve the management of cancer in the United States and in low and middle-income countries (LMICs). My graduate work focused on developing optical systems and automated algorithms to image tumor margins during surgery to improve the accuracy of cancer excision and reduce the likelihood that a patient needs to return for re-excision surgery. I investigated a variety of fluorescent stains, microscopy systems, model systems (including pre-clinical and clinical specimens), and algorithms, which resulted in 9 peer-reviewed publications (5 first author). My postdoctoral research has primarily focused on developing diagnostic and therapeutic technologies to better manage cervical pre-cancer in a global health context. First, I worked with a multidisciplinary and multicultural team to develop the Pocket colposcope, a low-cost, portable device to screen women for cervical pre-cancer at the primary care setting. We found that our device performs comparably to a standard-of-care colposcope through clinical studies at Duke University Medical Center and Liga Contra el Cancer in Perú, which resulted in 3 peer-reviewed publications (2 first author) and one patent. Second, I am developing a low-cost therapy to treat women who are diagnosed with cervical pre-cancer in a single visit to close the gap between diagnosis and treatment. The therapy leverages ethanol, a widely-used ablative agent, in combination with ethyl cellulose to form a gel in tissue to dramatically increase efficacy. We have demonstrated that ethyl cellulose-ethanol led to complete regression of oral tumors in a hamster cheek pouch model. In September 2018, I received a K99/R00 award from the NCI to optimize the delivery of ethyl cellulose-ethanol to treat cervical pre-cancer.

Education: Ph.D. in Biomedical Engineering, 2015, Duke University M.S. in Biomedical Engineering, 2013, Duke University B.S. in Bioengineering, 2009, Rice University

Research/Work Experience: Postdoctoral Associate, Duke University (Nimmi Ramanujam, David Katz), 2015 – present Research Assistant, Duke University (Nimmi Ramanujam), 2009 – 2015 Global Health Technologies Intern, Lesotho Baylor Pediatric AIDS Clinic (Maria Oden, Rebecca Richards-Kortum), 2008 Research Assistant, Rice University (Rebecca Richards-Kortum), 2007

Selected Publications: 1. Mueller J, Lam C, Dahl D, Asiedu M, Krieger M, Bellido Y, Kellish M, Peters J, Erkanli E, Ortiz E, Muasher L, Taylor P,S chmitt J, Venegas G, Ramanujam N. Portable Pocket colposcopy performs comparably to standard-of-care clinical colposcopy using acetic acid and Lugol’s iodine as contrast mediators: an investigational study in Perú. BJOG: An International Journal of Obstetrics and Gynaecology, 2018, 125(10): 1321-1329. 2. Morhard R, Hu F, Barrero Castedo B, Madonna M, Mueller J, Katz D, Ramanujam N. Optimization of Ethanol Ablation for Treatment of Precancerous Cervical Lesions in Resource-Limited Settings. Scientific Reports, 2017, 7: 8750. 3. Mueller J*, Dobbs J*, Krishnamurthy S, Shin D, Kuerer H, Yang W, Ramanujam N, Richards-Kortum R. Micro- anatomical quantitative optical imaging: towards automated assessment of breast tissues. Breast Cancer Research, 2015, 17: 105. 4. Mueller J, Fu H, Mito J, Whitley M, Chitalia R, Erkanli A, Dodd L, Cardona D, Geradts J, Willett R, Kirsch D, Ramanujam N. A quantitative microscopic approach to predict local recurrence based on in vivo intraoperative imaging of sarcoma tumor margins. International Journal of Cancer, 2015, 137(10): 2403-12.

Awards/Honors: Pathway to Independence Award (K99/R00), National Cancer Institute, National Institutes of Health, 2018-2023 Rice 360° Leadership Award, Rice 360° Institute for Global Health, Rice University, 2019 John T. Chambers Scholarship, Fitzpatrick Institute for Photonics (FIP), Duke University, 2013-2015 McChesney Fellowship in Biomedical Engineering, Department of Biomedical Engineering, Duke University, 2010-2011 Outstanding Service Bioengineering Departmental Award, Department of Bioengineering, Rice University, 2009 Clinton Global Initiative University Outstanding Commitment Award, Awarded by former President Bill Clinton for diagnostic Lab-in-a-Backpack research, Tulane University, 2008 BARIA R. MUTLU, PhD Center for Engineering in Medicine, Massachusetts General Hospital / Harvard Medical School, Boston, MA [email protected]

Research Overview: Cells contain a wealth of information as clinical biomarkers, and as research tools for studying physiological and pathological processes. My research focuses on developing systems for physical and chemical perturbation of cells in engineered microenvironments, for extracting information either by direct observation, or generating target-enriched samples for subsequent functional studies or omics analyses. Specifically, I am interested in developing micro-/nano-fluidic systems for the characterization, isolation, and imaging of circulating cells and cell complexes from peripheral blood. Towards this goal, I will be initially focusing on three research topics. 1) Circulating tumor cell (CTC) cluster biophysics: CTC clusters have drastically elevated metastatic potential compared to single CTCs. Thus, characterization of the biophysics of their cell-cell adhesion in flow conditions can reveal the underlying mechanisms leading to their increased tumorigenicity, and would enable rapid evaluation of drugs that can disrupt clusters under physiological stresses for antimetastatic therapies. 2) High-throughput sorting and analysis of submicron blood components: “Oscillatory inertial microfluidics” enables high-throughput, hydrodynamic manipulation of submicron particles in microchannels. Leveraging this technological advancement, I will develop technologies for label-free isolation of blood components such as: (i) Bacteria, for rapid identification of pathogens in septic patients, and (ii) Tumor-shed extracellular vesicles, for minimally invasive cancer diagnosis and prognosis. 3) Precision inertial microfluidics for next-generation flow cytometry applications: Imaging flow cytometry is one of the most ubiquitously employed and useful tools in life sciences, bioengineering and clinical research. Precision inertial microfluidics will improve this technology by highly sensitive cell positioning in a deterministic microfluidic environment, to enable acquisition of multiple time-series images of cells at different imaging orientations. This will allow dynamic measurements from cells, and construction of 3D cell images. Overall, my goal is to establish a research program with core competencies in: Cell biophysics, Micro-/nano-fluidics and Bioinstrumentation systems with the ultimate purpose of advancing the way we study, diagnose and treat diseases.

Education: PhD, Mechanical Engineering, 2016, University of Minnesota, Minneapolis, MN MSc, Mechanical Engineering, 2010, Middle East Technical University, Ankara, Turkey BSc, Mechanical Engineering, 2007, Middle East Technical University, Ankara, Turkey

Research/Work Experience: Postdoctoral Research Fellow, 2016-present, Massachusetts General Hospital / Harvard Medical School, Center for Engineering in Medicine, Boston, MA Graduate Research Assistant, 2010-2016, University of Minnesota, Biostabilization and Bioencapsulation Lab, Minneapolis, MN Graduate Research Assistant, 2007-2010, Middle East Technical University, Dynamic Systems and Controls Lab, Ankara, Turkey

Selected Publications: Mutlu B.R., Edd J.F., Toner M., “Oscillatory inertial focusing in infinite microchannels”, Proceedings of the National Academy of Sciences, 2018, 115 (30), 7682-7687 Mutlu B.R.*, Smith K.C.*, Edd J.F., Nadar P., Dlamini M., Kapur R., Toner M., “Non-equilibrium Inertial Separation Array for Highthroughput, Large-volume Blood Fractionation”, Scientific Reports, 2017, 7 (1), 9915. (*Co-first authored) Dietsche C.*, Mutlu B.R.*, Edd J.F., Koumoutsakos P., Toner M., “Dynamic Particle Ordering in Oscillatory Inertial Microfluidics”, Microfluidics and Nanofluidics, 2019, 23 (6), 83 (*Co-first authored) Mutlu B.R., Sakkos J.K., Yeom S, Wackett L.P., Aksan A., “Silica ecosystem for synergistic biotransformation”, Scientific Reports, 2016, 6, 27404. Mutlu, B. R., Yeom S., Wackett, L. P., Aksan, A, “Modelling and optimization of a bioremediation system utilizing silica gel encapsulated whole-cell biocatalyst”, Chemical Engineering Journal, 2015, 259: 574-580. Mutlu, B. R., Yeom S., Tong, H.-W., Wackett, L. P., Aksan, A, “Silicon alkoxide cross-linked silica nanoparticle gels for encapsulation of bacterial biocatalysts”, Journal of Materials Chemistry A, 2013, 1 (36), 11051-11060. Mutlu, B. R., Hirschey K., Wackett, L. P., Aksan, A, “Long-term preservation of silica gel encapsulated bacterial biocatalysts by desiccation”, Journal of Sol-Gel Science and Technology, 2015, 74 (3), 823-833.

Awards/Honors: Tosteson Award, Massachusetts General Hospital, 2019-2020 Doctoral Dissertation Fellowship, University of Minnesota, 2015-2016 Minnesota Discovery, Research and Innovation Fellowship, Biotechnology Institute / University of Minnesota, 2013-2015 DIANE LaVERN-LYGETTE NELSON, PhD Chemical Engineering, Carnegie Mellon University, 5000 Forbes Ave, Doherty 1107, Pittsburgh, PA, 15221 [email protected]

Research Overview: My research at Carnegie Mellon University investigated the benefits of water-in-perfluorocarbon reverse emulsions to improve the traditional treatment methods of lung diseases. Liquid perfluorocarbons (PFC) function like air, providing a medium for gas exchange; and as a delivery vehicle, enabling deeper lung penetration of delivered drugs. During my doctoral research in Biomedical Engineering, I optimized the formulation of antibiotic-loaded emulsions such that they overcame the quick transport of antibiotic away from the lung. This research led to the first antibiotic-in PFC emulsion to reduce pulmonary bacterial load in a rat respiratory infection model. Furthermore, I demonstrated that growth factors encapsulated into PFC emulsions maintain biological activity. For my postdoctoral research in Chemical Engineering, I engineered reverse PFC emulsions with controlled drug release profiles that would cater to specific therapeutic windows or disease states. I have determined that the limiting step in drug mass transfer is droplet coalescence with the receiving phase, which is strongly dependent on surfactant concentration. My research experiences uniquely qualify me to lead a research program that ranges from characterization to animal testing of complex fluids for biological applications of material encapsulation and delivery.

Education: -PhD, Biomedical Engineering, Carnegie Mellon University, May 2018 -MS, Colloids, Polymers, and Surfaces, Carnegie Mellon University, May 2018 -BS, Biomedical Engineering, Columbia University, May 2013 -BA, Mathematics, Georgetown University, May 2012

Research/Work Experience: Carnegie Mellon University -Postdoctoral Fellow (2018 – Present), Department of Chemical Engineering, Advisor: Robert Tilton, PhD -Graduate Research Assistant (2013 – 2018), Department of Biomedical Engineering, Advisor: Keith Cook, PhD Columbia University, 2012 – 2013 -Undergraduate Research Assistant, Department of Biomedical Engineering, Advisor: Samuel Sia, PhD

Selected Publications: 1. DL Nelson, L Chen, L Nistico, RA Orizondo, ML Fabiilli, KE Cook, "The Role of Convective Tobramycin Transport on Bacterial Biofilm Killing by Tobramycin-Loaded Reverse Perfluorocarbon Emulsions", Colloids and Surfaces B: Biointerfaces, in review. 2. DL Nelson, Y Zhao, ML Fabiilli, KE Cook, "In Vitro Evaluation of Lysophosphatidic Acid Delivery via Reverse Perfluorocarbon Emulsions to Enhance Alveolar Epithelial Repair", Colloids and Surfaces B: Biointerfaces (2018), Volume 169: 411-417, doi:10.1016/j.colsurfb.2018.05.037. 3. RA Orizondo, DL Nelson, ML Fabiilli, KE Cook, "Effects of Fluorosurfactant Structure and Concentration on Drug Availability and Biocompatibility in Water-in-Perfluorocarbon Emulsions for Pulmonary Drug Delivery", Colloid and Polymer Science (2017), Volume 295: 2413-2422, doi: 10.1007/s00396-017-4216-4.

Awards/Honors: -Burroughs Welcome Fund Postdoctoral Enrichment Program Fellowship, 2019 – 2022 -Burroughs Welcome Fund Collaborative Research Travel Grant, 2019 – 2020 -Presidential Postdoctoral Fellowship, Carnegie Mellon University, 2018 – 2020 -STAT Wunderkind, 2018 -Three Minute Thesis (3MT) Champion, Carnegie Mellon University, 2017 -NIH/NHLBI Graduate Research Supplement, 2016 – 2018 -Respiratory Drug Delivery Conference, Reviewer's Choice Award, 2016 -Dowd Fellowship, Carnegie Mellon University, 2015 – 2016 -Tau Beta Pi, Engineering Honor Society, 2012 CHRISTINE M. O’BRIEN, PhD Radiology, Washington University in St. Louis, 4515 McKinley Ave., Room 2207, St. Louis, MO, 63119 . [email protected]

Research Overview: I am a Biomedical Engineer focused on using my unique skill in non-invasive optics and imaging methods to solve global problems in Women’s Health. During my PhD, I developed point-based label-free optical spectroscopy as a tool to investigate how the main biochemical contributors to cervix tissue change during pregnancy in a 60 patient human trial as well as mouse models of preterm birth. In addition, I designed, built, and tested a speculum-free optical tool that significantly reduces challenges in clinical translation for optical monitoring of the cervix during pregnancy. During this time, I also completed a global health certificate including a practicum project conducting gastric cancer screening using optical spectroscopy in a rural Honduran hospital with high risk patients, which I funded with a pilot grant. In my postdoctoral work, I am extending my point- based spectroscopy knowledge to optical imaging projects that will aid in the diagnosis, intraoperative detection, and margin delineation of breast cancer. In the first project, I’ve employed a novel thermal imaging technique which reveals thermal properties oftissue at high spatial resolution, with cancerous areas having unique responses compared to healthy tissue. For my second project, I have devised a fluorescence depth determination approach for use in fluorescence guided breast conserving surgery and tumor margin delineation. In summary, a central theme to my work is the development of translational, point-of- care, optical technologies that meet Women’s Health needs. I am motivated by unsolved medical problems and healthcare inequities in Women’s Health, in high and low resource areas alike. In my future laboratory, I plan to capitalize on my passion for Global Women’s Health and expand upon my technical foundation to develop technologies that help women while addressing practical barriers to ensure the technologies reach patients who need them most.

Education: -Ph.D., Biomedical Engineering, 2017, Vanderbilt University -Teaching Certificate, Global Health Certificate -B.S., Bioengineering, 2011, University of Missouri

Research/Work Experience: -W. M. Keck Postdoctoral Fellow, Washington University in St. Louis School of Medicine, Department of Radiology, January 2018 – present, Advisor: Samuel Achilefu, Ph.D. - Graduate Fellow, Vanderbilt University, Department of Biomedical Engineering, 2011-17; Advisors: Anita Mahadevan- Jansen, Ph.D. and Jeff Reese, M.D. - Undergraduate Researcher, University of Missouri, Department of Bioengineering, 2008-11, Advisor: John Viator, Ph.D. - NSF REU Fellow, University of Cincinnati, Department of Molecular and Cellular Physiology, 2008

Selected Publications: -CM O’Brien, KJ Cochran, L Masson, M Goldberg, E Marple, KA Bennett, J Reese, JC Slaugter, JM Newton, A Mahadevan- Jansen, “Development of a visually guided Raman spectroscopy probe for cervical assessment during pregnancy,” J Biophotonics, (2018). -CM O’Brien*, E Vargis*, A Rudin, JC Slaughter, KA Bennett, J Reese, A Mahadevan-Jansen, “In vivo Raman spectroscopy for biochemical monitoring of the cervix during pregnancy,” Am J Obstet Gynecol, 218(5), (2018). *Equally contributing authors. -L Masson, CM O’Brien, IJ Pence, JL Herington, J Reese, T van Leeuwen, and A Mahadevan-Jansen, “Dual Excitation Wavelength System for Combined Fingerprint and High Wavenumber Raman Spectroscopy,” Analyst, 143, (2018). -CM O’Brien, JL Herington, N Brown, IJ Pence, BC Paria, JC Slaughter, J Reese, A Mahadevan-Jansen, “In vivo Raman spectral analysis of impaired cervical remodeling in a mouse model of delayed parturition” Sci Rep, 7(1), (2017). -CM O’Brien, KD Rood, K Bhattacharyya, T DeSouza, S Sengupta, SK Gupta, J Mosley, BS Goldschmidt, N Sharma, JA Viator, “Capture of circulating tumor cells using photoacoustic flowmetry and two phase flow” J Biomed Opt 17(6), (2012).

Awards/Honors: W. M. Keck Postdoctoral Fellowship, 2018-2019 Outstanding poster award at SCIX 2017, 2017 Vanderbilt Laboratories for Innovation in Global Health Technologies (LIGHT) Pilot Grant, 2016-2017 Vanderbilt Global Health Case Competition Finalist, 2016 Philanthropic Education Organization (PEO) Scholarship Recipient, 2015-2016 National Consortium for Pediatric Device Innovation Finalist, 2015 NIH R01 HD081121, significant contributions to the research strategy, 2014-2019 SPIE Scholarship in Optics and Photonics, 2012, 2014 National Science Foundation Graduate Research Fellowship, 2011-2014 MOLLY E. OGLE, PhD Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, GA, 30332 [email protected]

Research Overview: My postdoctoral research has primarily explored strategies to promote healing of musculoskeletal injuries through biomaterial- and mmesenchymal stem cell- mediated control of recruitment, localization, and activity of endogenous immune cell populations and expansion of microvascular networks. Biomaterial implants have tremendous power to modulate the activities and recruitment dynamics of immune and progenitor cell populations through spatially localized presentation of bioactive signaling molecules, adhesive ligands, and structural elements. I have utilized glycosaminoglycan (GAG)-based biomaterials to facilitate tunable affinitybased loading and release of many chemokines and growth factors that can modulate cell behavior to improve metrics of repair. My studies not only strive to develop translational biomaterials to promote wound healing, but also to understand the complexities of the innate immune response to injury and how to best tune the immune response to improve healing particularly in the context of an implanted material. During the course of my work, I have developed methods and built expertise in intra-vital tracking of regenerative immune cell sub-populations to sites of injury, particularly within the dorsal skinfold window chamber model and the mouse cranial defect model. Overall, my research has maintained a focus on understanding the mechanisms of fundamental endogenous repair processes in order to apply this knowledge to pathological systems and design criteria for novel biomaterials. The goal for my future research group will be to pair this work with my graduate expertise in neurological disease to examine ways that biomaterials can support repair in central and peripheral nervous system disorders. My background in vascular remodeling, harnessing endogenous mechanisms of repair, cell signaling networks, intra-vital cell tracking, and immune cell phenotyping provide substantial expertise to lay a foundation for my long-term research goals in utilizing endogenous cellular protective pathways for translational therapeutic design.

Education: Emory University, Atlanta, GA 2012; PhD Biochemistry, Cell, Developmental Biology University of Virginia, Charlottesville, VA 2004; BS Biology

Research/Work Experience: -2017-present: Georgia Institute of Technology, Department of Biomedical Engineering, Postdoctoral Fellow, Mentor: Dr. Johnna Temenoff -2012-2018: Georgia Institute of Technology, Post-doctoral Fellow, Mentor: Dr. Edward Botchwey -2008-2012: Emory University, Graduate Research Assistant, Mentor: Dr. Ling Wei

Selected Publications: -Ogle ME, Krieger JR, McFaline-Figueroa J, Temenoff JS, Botchwey EA. Dual-Affinity Heparin Hydrogels Achieve Localized Immunomodulation And Enhance Microvascular Remodeling. ACS Biomaterials Sci & Eng. (2017) -Olingy CE*, San Emeterio CL*, Ogle ME, et al. Non-classical monocytes are biased progenitors of wound healing macrophages during soft tissue injury. , Scientific Reports. (2017) -Ogle ME, et al. Monocytes and macrophages in tissue repair: Implications for biomaterial design. Exp Biol Med. (2016) -Krieger JR*, Ogle ME*, et al. Spatially localized recruitment of anti-inflammatory monocytes by SDF-1α-releasing hydrogels enhances microvascular network remodeling. Biomaterials. (2015) *co-first authorship -Ogle ME, et al. Engineering in vivo gradients of sphingosine-1-phosphate receptor ligands for localized microvascular remodeling and inflammatory cell positioning. Acta Biomaterialia. (2014) -Awojoodu AO, Ogle ME, et al. Sphingosine-1-phosphate receptor 3 regulates recruitment of anti-inflammatory monocytes to microvessel during implant arteriogenesis. PNAS (2013). -Ogle ME, Gu X, Espinera AR, and Wei L. Inhibition of prolyl hydroxylases by dimethyloxaloylglycine after stroke reduces ischemic brain injury and requires hypoxia inducible factor-1. Neurobiology of Disease. (2012)

Awards/Honors: -BMES Poster Award 2016 -Best trainee oral presentation, Biomaterials & Tissue Engineering Gordon Research Seminar: 2015 -Southeastern Regional Lipid Conference Research Travel Award: 2014 -Gandy-Diaz Senior Teaching Fellow: 2012, 2013 -Emory Crawford W. Long Excellence in Research Award: 2010, 2011 LAURA L. OSORNO, PhD Biomimetic & Biohybrid Materials, Biomedical Devices, & Drug Delivery Laboratories, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ, 08028 - [email protected]

Research Overview: My doctoral research involves engineering, designing, and characterizing a state-of-the-art dual delivery platform. The platform consists of an injectable, biodegradable optically clear, and in-situ forming nanogel for the extended and controlled release of DNA-based ocular therapeutic nanospheres over a month. This dual delivery platform will potentially become a prophylactic treatment for the early prevention of secondary cataracts. My future research will focus on novel biomaterials engineering, therapeutic drug delivery, pharmaceutical engineering, and nanotechnology. My long-term research goals are: (i) to pioneer novel self-assembled, injectable biomaterials for the development of local and targeted nanocarriers for pain management, ocular, and immunology applications; (ii) to develop transdermal drug delivery systems to limit inflammation and prevent severe scarring during wound healing; (iii) to achieve understanding of the mechanobiology of dermal and neural cells in response to physical force, inflammation, and aging, including how these properties translate to serious medical conditions.

Education: • Rowan University, September 2019. PhD, Biomedical Engineering • New Jersey Institute of Technology, May 2015. MS, Biomedical Engineering • New Jersey Institute of Technology, May 2014. BS, Biomedical Engineering

Research/Work Experience: • Rowan University, 2015 – 2019, Graduate Research Assistant Biomedical Engineering Advisor: Mark E. Byrne, PhD, Founding Department Head and Professor • New Jersey Institute of Technology, 2014 – 2015, Graduate Research Assistant Biomedical Engineering; Advisor: Michael Jaffe, PhD, Professor • New Orleans University, Summer 2013, Undergraduate Research Intern, Chemical Engineering; Advisor: Matthew A. Tarr, PhD, Professor • Union County College, 2012 – 2015, Supervisor of Academic Tutoring in Mathematics

Selected Publications & National/International Conference Presentations (of 15): 1. Osorno L, George-Weinstein M, Byrne ME. Optically Clear, In-Situ Forming Biodegradable Nano-Carriers for Ocular Therapy, and Methods Using Same. Patent WO 2018/18398 A. 2. Osorno L, Maldonado DE, Whitener RJ, Brandley AN, Yiantsos A, Medina JDR, Byrne ME. Amphiphilic PLGA-PEG-PLGA Triblock Copolymer Nanogels Varying in Gelation Temperature and Modulus for the Extended and Controlled Release of Hyaluronic Acid. Journal of Polymer Sciences. Submitted (2019) 3. Osorno L, Jaffe M. Behavior of Deformed Fibroblasts Seeded on a Silicone Membrane under High Levels of Strain. Master’s Thesis, Biomedical Engineering Department. New Jersey Institute of Technology: Newark, New Jersey (2015) 4. U.S. Patil, L. Osorno, A. Ellender, C. Grimm, M. A. Tarr. Cleavable Ester-Linked Magnetic Nanoparticles for Labeling of Solvent-Exposed Primary Amine Groups of Peptides/Proteins. Journal of Data in Brief. 484: (2015)

Awards/Honors: • BMES Career Development Travel Award. Biomedical Engineering Society. Fall 2018. • Public Policy Institute for Rising Leaders Travel Award. American Institute for Medical and Biological Engineering (AIMBE). Fall 2017. • Founder of BME-Kindle Program. Rowan University. Summer 2017 – Present: As a Hispanic Engineer, this program was founded to educate and inspire high school students, especially underrepresented, in the field of biomedical engineering. SEUNGMAN PARK, PhD Whiting School of Engineering/Institute for NanoBioTechnology, Johns Hopkins University, 3400 N. Charles St., Latrobe 216B, Baltimore, MD, 21218 - [email protected]

Research Overview: My research focuses on studying 1) functionality assessment and mechanical characterization of biological systems, 2) treatment of diseases using the mechanics-based platform, and 3) understanding the underlying molecular/cellular mechanism of diseases. In addition, I am researching an essential biological question “how mechanical or biochemical cues in diseased or abnormal microenvironment affects cell function, behavior, and vice versa?” through quantitative analysis of biophysical observations. For the quantitative analysis, I am developing various tools for quantitating drag/traction forces, bond strength, and mechanical/viscoelastic/transport properties of cells, tissues and biomaterials. I envision a future where quantitative methods and tools enable new discoveries of basic/clinical science, thereby encouraging physicians and clinicians to detect or treat diseases more easily. During my PhD studies, my research interest had been focused on the development of functional engineered tissues (ETs) and effective ways for long-term storage of ETs by investigating the structure and functionality relationship. By measuring tissue viscoelastic properties, I discovered that the cytoskeletal structure and the use of CPAs were strongly correlated to the preservation of the post-thaw functional properties My past training has laid a solid foundation for my current efforts in developing functional engineered tissues and characterizing the mechanical behavior of various types of tissues or cells during my postdoctoral training at Johns Hopkins University. My extensive background in biomechanics and tissue engineering enabled me to quantitate drag forces exerted on cancer cells, characterize contractility (contraction force, period, and deformation rate) of cardiac tissues, and develop mechanical tools for measurement of viscoelastic properties from cellular to tissue level using microfluidics and indentation-based force sensor. I also aim to understand the underlying mechanism of cancer-induced immunosuppression and study the complex relationship between tissue microenvironment and cell behavior through regulation of signaling pathway, gene expression, and protein turnover.

Education: • Purdue University, December 2014. PhD, Mechanical Engineering • Pusan National University, February 2010. MS, Mechanical Engineering • Pusan National University, February 2008. BS, Mechanical Engineering

Research/Work Experience: 01/2016 - present: Postdoctoral Research Associate, Johns Hopkins University 07/2015 - 12/2015: Visiting Research Specialist, University of Illinois at Chicago 01/2015 - 06/2015: Postdoctoral Research Associate, Purdue University 08/2010 - 12/2014: Graduate Research Assistant, Purdue University

Selected Publications: 1. Seungman Park, Yoon Ki Joo, Yun Chen, “Versatile and High-throughput Force Measurement Platform for Dorsal Cell Mechanics” Scientific Reports, Accepted 2. Seungman Park, Jiaxiang Tao, Li Sun, Chen-Ming Fan, Yun Chen, “Economic, Modular and Portable Skin Viscoelasticity Measurement Device for in situ Longitudinal Studies” Molecules, 2019, 24(5), 907 3. Seungman Park, Cecillia Lui, Wei‐Hung Jung, Debonil Maity, Chin Siang Ong, Joshua Bush, Venkat Maruthamuthu, Narutoshi Hibino, Yun Chen, “Mechanical characterization of hiPSCs-derived cardiac tissues for quality control” Advanced Biosystems, 2018, 2(12), 1800251 4. Seungman Park, Angela Seawright, Sinwook Park, Craig Dutton, Fred Grinnell, Bumsoo Han, “Preservation of tissue microstructure and functionality during freezing by modulation of cytoskeletal structure” Journal of the Mechanical Behavior of Biomedical Materials, 2015. 45: p. 32-44.

Awards/Honors: • Maryland Stem Cell Research Foundation Postdoctoral Fellowship. 2018 - 2020. • NSF CBET Travel Award, ASME Summer Bioengineering Conference. 2011 • Scholarship, Brain Korea 21, Korea Research Foundation. 2008 - 2010 • Academic Excellence Scholarship (full tuition), Pusan National University. 2005 - 2007 JAY MILAN PATEL, PhD 1McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, PA, 19104, 2Corporal Michael J Crescenz VA Medical Center, Phiadelphia, PA, 19104 [email protected]

Research Overview: As an independent research investigator, I am interested in the role that cellular microenvironments play in governing musculoskeletal tissue repair and regeneration. Specifically, I wish to manipulate cell surroundings in order to control behavior and ultimately translate these findings towards providing macro-scale clinical utility, as they relate to orthopaedic scaffolds and therapeutics. During my doctoral dissertation work, I focused on the latter stages of this paradigm by developing a fiber-reinforced acellular meniscus scaffold and testing its preclinical efficacy in a large-animal model and functional biomechanics in a human cadaveric knee model. While not explored, I became intrigued by the behavior of cells recruited to this scaffold; thus, during my postdoctoral training, I shifted towards evaluating the micro- scale mechanobiology of cells and their response to their environment. Specifically, I studied the adhesion of progenitor cells to focal cartilage defects, and developed a novel biomaterial system that interdigitates with the damaged cartilage and provides a chemically and mechanically modified microenvironment to improve cellular attachment and mechano- sensation. Ultimately, these cells were directed to lay down robust extracellular matrix, and I am currently evaluating the ability of this approach to seal cartilage defects and prevent further articular deterioration. With these experiences in hand, I am excited to work with biologists, materials scientists, engineers, and clinicians in order to study and enhance cellular microenvironments in the context of orthopaedic tissue engineering and regenerative medicine, with the goal of taking novel therapeutic approaches from conceptualization to the clinic.

Education: 2017 Ph.D. in Biomedical Engineering, Rutgers University 2011 B.S. in Bioengineering, Rice University

Research/Work Experience: 2017- Postdoctoral Fellow, University of Pennsylvania 2016- Consultant, NovoPedics Inc. 2011-2017 Graduate Student Researcher, Rutgers University 2011 Process Development Intern, LifeCell Corporation 2008-2010 Undergraduate Research Assistant, Rice University

Selected Publications: [1] Patel JM, Dunn MG, Gatt CJ. “System and method for making personalized fibrocartilage implants.” International PCT Application No. WO2017095662 A1. June 8, 2017. [2] Patel JM, Ghodbane SA, Brzezinski A, Gatt CJ, Dunn MG (2018) Tissue-engineered total meniscal replacement using a fiberreinforced scaffold in a two-year ovine model. AJSM. 46(8): 1844-1856. [3] Patel JM, Saleh KS, Burdick JA, Mauck RL (2019) Bioactive Factors for Cartilage Repair and Regeneration: Improving Delivery, Retention, and Activity. Acta Biomateriala. 93: 222-238. [4] Martin AR, Patel JM, Zlotnick HM, Carey JL, Mauck RL (2019) Emerging Therapies for Cartilage Regeneration in Currently Excluded ‘Red Knee’ Populations. Nature Regenerative Medicine. 4(1): 1-12. [5] Patel JM, Wise BC, Bonnevie ED, Mauck RL (2019) A Systematic Review and Guide to Mechanical Testing for Articular Cartilage Tissue Engineering. Tissue Engineering Part C. Epub Ahead of Print. [6] Patel JM, Loebel C, Martin AR, Mauck RL, Burdick JA. “Systems for Targeted Tissue BioSealing or Repair.” US Patent Application No. US 62/697,252. July 12, 2019.

Awards/Honors: Career Development Award (CDA-1), Department of Veterans Affairs, 2019 Top Podium Presentation Award, Department of Orthopaedic Surgery, 2019 Excellence in Research Award, American Orthopaedic Society for Sports Medicine, 2018 New Investigator Recognition Award, Orthopaedic Research Society, 2018 Poster Finalist, J&J Engineering Showcase, 2016 Inaugural Willy’s Revolution Award, Rice University, 2011 COLIN D. PAUL, PhD Laboratory of Cell Biology, National Cancer Institute, 37 Convent Drive, Bldg 37, Room 2132, Bethesda, MD, 20892 [email protected] Research Overview: Metastasis describes the process where cancer cells move from a tumor to establish lesions in organs distinct from the primary site and represents a key clinical bottleneck, with five-year survival rates for patients with metastatic disease remaining under~30% for many cancers. My research program will elucidate how and why physical forces, especially those related to the topography of the microenvironment, influence the formation of metastatic tumors at secondary sites. My scientific background makes me uniquely qualified to carry out research at the interface of engineering and cancer cell biology. During my doctoral research with Dr. Konstantinos Konstantopoulos at Johns Hopkins University, I studied mechanisms of confined tumor cell migration using custom-engineered microfluidic devices. For my postdoctoral research with Dr. Kandice Tanner at the National Cancer Institute (NCI), I developed a zebrafish xenograft model of early metastatic dissemination to visualize the transition of circulating tumor cells from the bloodstream to target organs in vivo at single-cell resolution. As an independent investigator, I will combine these techniques to provide important information on the basic biology of cancer metastasis and identify druggable targets for metastatic disease. Biologically-inspired microfluidic models will be used to probe how environmental topography promotes cancer cell arrest and extravasation. Intravital imaging of cancer and immune cells during metastasis will concurrently be analyzed in environments of varying topographical complexity in zebrafish. Ultimately, this research will identify and modulate interactions between disseminating cancer cells and the physical microenvironment to target nascent secondary tumors before they grow to the point of impairing tissue function.

Education: • Ph.D., Chemical and Biomolecular Engineering, Johns Hopkins University (2015) • B.S., Chemical Engineering, B.S., Physics, University of Arkansas (2010)

Research/Work Experience: • National Cancer Institute (2015-present) Cancer Research Training Postdoctoral Fellow, Laboratory of Cell Biology; Advisor: Kandice Tanner, Ph.D. • Johns Hopkins University (2010-2015) Graduate Research Assistant, Chemical and Biomolecular Engineering; Advisor: Konstantinos Konstantopoulos, Ph.D.

Selected Publications: 1. C.D. Paul, K. Bishop, A. Devine, E.L. Paine, J.R. Staunton, S.M. Thomas, J.R. Thomas, A.D. Doyle, L. M. Miller Jenkins, N.Y. Morgan, R. Sood, and K. Tanner. Tissue architectural cues drive organ targeting of tumor cells in zebrafish. Cell Systems 9: 187-206 (2019) 2. C.L. Yankaskas, K.N. Thompson, C.D. Paul, M.I. Vitolo, P. Mistriotis, A. Mahendra, V.K. Bajpal, D.J. Shea, K.M. Manto, A.C. Chai, N. Varadarajan, A. Kontrogianni-Konstantopoulos, S.S. Martin, and K. Konstantopoulos. A microfluidic assay for the quantification of the metastatic propensity of breast cancer specimens. Nature Biomedical Engineering 3: 452-465 (2019) 3. C.D. Paul, A. Hruska, J.R. Staunton, H.A. Burr, K.M. Daly, J. Kim, N. Jiang, and K. Tanner. Probing cellular response to topography in three dimensions. Biomaterials 197: 101-118 (2019) 4. C.D. Paul*, A. Devine*, K. Bishop, Q. Xu, W.J. Wulftange, H. Burr, K.M. Daly, C. Lewis, D.S. Green, J.R. Staunton, S. Choksi, Z.-G. Liu, R. Sood, and K. Tanner. Human macrophages survive and adopt activated genotypes in living zebrafish. Nature Scientific Reports 9: 1759 (2019) *These authors contributed equally 5. C.D. Paul, P. Mistriotis, and K. Konstantopoulos. Cancer cell motility: lessons from migration in confined spaces. Nature Reviews Cancer 17: 131-140 (2017) 6. C.D. Paul, D.J. Shea, M.R. Mahoney, A. Chai, V. Laney, W.-C. Hung, and K. Konstantopoulos. Interplay of the physical microenvironment, contact guidance, and intracellular signaling in cell decision making. The FASEB Journal 30: 2161-2170 (2016)

Awards/Honors: • Best Oral Presentation Award, 2019 Center for Cancer Research Fellows and Young Investigators Colloquium (2019) • Finalist, National Cancer Institute Outstanding Postdoctoral Fellow Award (2018) • National Cancer Institute Director’s Innovation Award (2017) • Johns Hopkins Chemical and Biomolecular Engineering Graduate Student Award (2015) • Biomedical Engineering Society Graduate Design and Research Award (2013) • Johns Hopkins University Schwarz Fellowship (2010-2015) • National Science Foundation Graduate Research Fellowship (2011-2014) JUDE M. PHILLIP, PhD Medicine, Weill Cornell Medicine, 1300 York Avenue, C640A, New York, New York, 10065 [email protected]

Research Overview: My research focuses on applying engineering principles to solving key biological questions, primarily in the areas of cancer and aging. During my undergraduate studies, my early interested in research afforded me two profound research opportunities—working with Alexander Couzis at CCNY and a summer with Richard Zare at Stanford University—which contributed to shaping my career path of translational bioengineering. As a doctoral student at Johns Hopkins University under the guidance of Denis Wirtz, I co-developed an automated high-throughput cell phenotyping platform to probe morphological changes of cells as functions of perturbations. Later, combining this platform with other biophysical profiling modalities, I developed a cell-based approach to determine the cellular biological age of healthy individuals using primary dermal fibroblasts. This work directly led to a publication in Nature Biomedical Engineering, and successfully securing a U01 grant from the National Institutes of Aging (NIA), of which I am a Co-PI. This is an important development since it potentially impacts clinical decision making with regards to stratification and intervention approaches to improve the functional healthspan of individuals. With a desire to enhance my abilities in identifying key questions in medicine that could benefit from engineering solutions, I moved to Weill Cornell Medicine to learn alongside Physician-Scientists: Leandro Cerchietti and Ari Melnick. Here, I am pioneering a study to elucidate the role of microenvironmental stress tolerance on extracellular matrix composition and architecture, and stroma-immune crosstalk. With these experiences I am developing a unique career niche with an overarching vision seeks to improve human healthspan and disease outcomes by developing technologies and other engineered solutions for pre-clinical applications.

Education: • Johns Hopkins University, October 2015, PhD., Chemical and Biomolecular Engineering • City College of New York, May 2010, B.Eng., Chemical Engineering; Magna Cum Laude

Research/Work Experience: • Postdoctoral Associate, Weill Cornell Medicine, Hematology and Oncology, 2016-present, PIs: Leandro Cerchietti, MD. & Ari Melnick, MD. • PhD candidate, Johns Hopkins University, Chemical and Biomolecular Engineering, 2010-2015, PI: Denis Wirtz, PhD. • HHMI EXROP Visiting student researcher, Stanford University, Chemistry, 2008 (summer), PIs: Richard Zare, PhD. and Gunilla Jacobson, PhD. • Student researcher, City College of New York, Chemical Engineering, 2007-2010, PI: Alexander Couzis, PhD.

Selected Publications: • T.M. Fernando*, R. Marullo*, B. Pera, J.M. Phillip, […], A. Melnick, L. Cerchietti, BCL6 evolved to enable stress tolerance in vertebrates and is required by cancer cells to adapt to stress, 2019, Cancer Discovery • J.M. Phillip, P-H. Wu, D. Gilkes, […], J. Walston, D. Wirtz, Biophysical and biomolecular determination of cellular age in humans, 2017, Nature Biomedical Engineering • P-H. Wu*, J.M. Phillip*, S.B. Khatau, W-C. Chen, […], D. Wirtz, Evolution of cellular morpho-phenotypes in cancer metastasis, 2015, Scientific Reports • J.M. Phillip*, I. Aifuwa*, J. Walston, D. Wirtz, The Mechanobiology of Aging, 2015, Annual Review of Biomedical Engineering • Y. Yu*, S. Gaillard*, J.M. Phillip, […], D. Wirtz, T-L. Wang, I-M. Shih, Inhibition of SYK potentiates paclitaxel- induced cytotoxicity in ovarian cancer cells through stabilizing microtubules, 2015, Cancer Cell

Awards/Honors: • National Society for Black Engineers (NSBE)—Biomedical Engineering Society Travel Award, 2019 • Biomedical Engineering Society (BMES) Career Development Award, 2018 • American Association for Cancer Research (AACR) Scholar-in-Training Award, 2018 • Predoctoral Fellow, National Cancer Institute—Cancer Nanotechnology Training Center, 2010-13 • Inducted Member, Tau Beta Pi—National Engineering Honor Society, 2008 • Howard Hughes Medical Institute—Exceptional Research Opportunity Program Award (EXROP), 2008 ANA MARIA PORRAS, PhD Biomedical Engineering, Cornell University, 290 Kimball Hall, Ithaca, NY, 14850 . [email protected] Research Overview: The Porras Biomaterials and Microbe Engineering (BIOME) lab will develop in vitro models to understand how microorganisms interact with their microenvironment to regulate human health and disease. Much remains to be elucidated regarding the specific interactions between microbes and their human hosts. This is particularly true in the context of the ECM, which provides important structural and biochemical cues for the development, homeostasis, and eventual onset and progression of disease in all human tissues. To address this need, I am currently engineering in vitro platforms to study the interplay between commensal bacteria and human gut ECM in inflammatory bowel disease (IBD). During my postdoctoral training, I have uncovered that multiple commensal bacterial strains have the ability to degrade components of the gut ECM including those in the basement membrane. I am now designing ECM-based hydrogels to study how bacterial modifications of the matrix impact macrophage and fibroblast behavior to influence IBD progression. My research program will continue to build on this work to develop in vitro models of disease to study host-microbe interactions. Through a combination of biomaterials and -omics approaches, we will engineer models of cardiovascular and gastrointestinal diseases to understand how commensal, opportunistic, and pathogenic microorganisms interact with the ECM to drive pathological outcomes in diseases like IBD, colorectal and stomach cancer, and Chagas disease. My research program will incorporate my background in (1) tissue engineered in vitro and ex vivo models of disease, (2) bacterial cultures, sequencing technologies and germ-free mouse models for the study of the human microbiome, and (3) immunology in the context of both inflammation and infectious disease. During my doctoral training, I used natural biomaterials and ex vivo gene delivery to design models that mimicked the features of the extracellular matrix (ECM) in early calcific aortic valve disease, a disease for which there is currently no treatment. Through these disease-inspired platforms, I identified a previously unknown cascade of pathological events that culminated in the initiation of an inflammatory response. To expand the breadth of my work, I accepted a postdoctoral position to study the human gut microbiome, where I have been exploring differences in the microbiome between populations in developed and developing countries. Using germ-free mouse models, I established that these geographic differences in gut microbiota composition have an effect on susceptibility to infection through the mucosal T helper 17 pathway. I seek to leverage my diverse and unique training in both tissue engineering and the gut microbiome to pursue a highly interdisciplinary line of research that contributes to the identification of microbial contributors to human health and disease.

Education: 2016 | Ph.D. Biomedical Engineering, University of Wisconsin-Madison 2015 | Delta Certificate in Teaching and Learning, University of Wisconsin-Madison 2013 | M.S. Biomedical Engineering, University of Wisconsin-Madison 2011 | B.S. Biomedical Engineering with High Honors, University of Texas at Austin

Research/Work Experience: 2017 – Present | Postdoctoral Fellow, Cornell University (Advisor: Ilana Brito) 2011-2016 | Graduate Research Fellow, University of Wisconsin-Madison (Advisor: Kristyn Masters) 2013-2014 | Teaching as Research Intern, University of Wisconsin-Madison (Advisors: Susan Nossal and Amihan Huesmann) 2008-2011 | Undergraduate Research Assistant, University of Texas at Austin (Advisor: Christine Schmidt)

Selected Publications: 1. Porras AM, Brito IL. The internationalization of human microbiome research. Current Opinion in Microbiology, Accepted, 2019. 2. Porras AM et al. Creation of Disease-Inspired Biomaterial Environments to Mimic Pathological Events in Early Calcific Aortic Valve Disease. Proceedings of the National Academy of Sciences, 2017. 3. Porras AM et al. Robust Generation of Quiescent Porcine Valvular Interstitial Cell Cultures. Journal of the American Heart Association, 2017. *Denotes equal contribution by authors 4. Porras AM et al. Engineering Approaches to Study Fibrosis in 3-D in Vitro Systems. Current Opinions in Biotechnology, 2016.

Awards/Honors: Cornell Presidential Postdoctoral Fellowship (2019-2021) Postdoc Achievement Award for Excellence in Community Engagement, Cornell (2018) American Heart Association Predoctoral Fellowship (2015-2016) Patricia Heard Outstanding Student Educator Award, UT-Austin (2011) TONGCHENG QIAN, PhD Morgridge Institute, University of Wisconsin-Madison, 330 N. Orchard St, Madison, WI, 53715 [email protected] Research Overview: My research at Peking University and the University of Wisconsin-Madison has been focused on biomaterials and tissue engineering. During the first two years of my Ph.D. program at Peking University, I focused on material science, conducting research related to surface modification by surface-initiated polymerization (SIP). During my third and fourth year of my Ph.D. program, I received a scholarship from the Chinese government, and I went to the University of Illinois at Urbana-Champaign to conduct research in live cell imaging by integrating micropattern technology with fluorescence resonance energy transfer technique. During my postdoctoral training at the University of Wisconsin- Madison, I have been focused on stem cell engineering, including studying the mechanisms of the blood brain barrier (BBB) development under the control of microenvironments, differentiation of BBB endothelial cells, and generating heart organoids for disease modeling with stem cell-derived heart cells, in which there are tremendous opportunities for disease modeling, drug discovery and clinical application. To be a part of an innovative, creative, and truly helpful field, my future research will be focused on tissue engineering and tissue regeneration from stem cells for disease modeling and drug screening.

Education: ●Peking University, July 2011. Ph.D., Biomechanics and Medical Engineering ●Ningbo University, July 2005. Bachelor of Medicine

Research/Work Experience: ●University of Wisconsin-Madison, 2017.11-present, Assistant Scientist, Morgridge Institute, Advisor: Melissa Skala, Ph.D. ●University of Wisconsin-Madison, 2014.1 -2017.11 Research Associate, Chemical and Biological Engineering, Advisor: Sean Palecek, Ph.D. & Eric Shusta, Ph.D. ●SinoMD Diagnostics, Beijing, China, 2013.1 -2013.12, Scientist ●Chinese Academy of Sciences, 2011.7 – 2013.1 Research Associate, Institute of Zoology, Beijing, China, Advisor: Baoyang Hu, Ph.D. ●Board of Health, Xiangshan County, Zhejiang, China, 2003–2006, Civil Servant

Selected Publications: 1. Qian, T., Li, Y., Wu, Y., Zheng, B., Ma, H. Superhydrophobic poly(dimethylsiloxane) via surface-initiated polymerization with ultralow initiator density. Macromolecules, 2008, 41 (18): 6641–6645 2. Qian, T., Wang, Y. Micro/nano-fabrication technologies for cell biology. Medical & Biological Engineering & Computing, 2010, 48(10): 1023-1032 3. Qian, T., Lu, S. Ma, H., Fang, J., Zhong, W., Wang, Y. FRET imaging of calcium signaling in live cells in the microenvironment. Integrative Biology. 2013, 5(2): 431-8 4. Zhuo, Y., Qian, T. (Equal contribution), Wu, Y., Seong, J., Gong, Y., Ma, H., Lu, S., Wang, Y. Subcellular and dynamic coordination between Src activity and cell protrusion in microenvironment. Scientific Reports 5, Article number: 12963 (2015) 5. Qian, T., Shusta, E., Palecek. S. Advances in microfluidic platforms for analyzing and regulating human pluripotent stem cells. Current Opinion in Genetics & Development. 2015 (34): 54-60. 6. Qian, T., Maguire, S., Bao, X., Olson, W., Shusta, E., Palecek, S. Directed differentiation of human pluripotent stem cells to blood-brain barrier endothelial cells. Science Advances. 2017;3: e1701679 7. Qian, T., Maguire, S., Canfield, S., Bao, X., Olson, W., Panzer, S., Shusta, E., Palecek, S. Directed Differentiation of Human Pluripotent Stem Cells to Podocytes under Defined Conditions. Scientific reports, 2019; 9 (1), 2765

Awards/Honors: ●National Scholarship, Ministry of Education of China. 2008-2010 ●ISCT - SCRMC Career Development Award, 2019 FEINI QU, VMD, PhD Orthopaedic Surgery, Washington University in St. Louis, 4515 McKinley Ave, Box 8233, St. Louis, MO, 63108 [email protected]

Research Overview: Extensive interdisciplinary training in tissue engineering, biomaterials, drug delivery, mechanobiology, and veterinary medicine has prepared me for a research career in musculoskeletal regenerative medicine. My long-term career goal is to develop and translate therapeutics that promote tissue repair and regeneration after injury, especially with respect to the bone and connective tissues of limbs and joints. My research focuses on (1) understanding how cells coordinate repair after injury and (2) modulating the cellular response and wound microenvironment to guide tissue repair. For my doctoral work, I developed tunable nanofibrous scaffolds that sequentially delivered multiple therapeutic biofactors to tears of the knee meniscus to improve endogenous cellular repair in vivo. Using a novel tissue-based 3D migration assay, I demonstrated that targeted reprogramming of the microscale structure and mechanics of the extracellular matrix enhances repair by facilitating cell migration to the wound site. To advance translational studies in large animal models of joint injury, I created a wearable device that evaluates joint kinematics using a sensor and magnet system in a noninvasive and unsupervised manner (patent pending). As a postdoctoral fellow, I am using molecular, bioinformatic, and transgenic techniques to elucidate the dynamic transcriptional landscape underlying murine digit tip regeneration. My work identified a gene regulatory network that recapitulates aspects of limb development, as well as several progenitor cell populations that mediate the regrowth of bone and fibrous tissues after digit amputation. In my laboratory, we will use these translational platforms to study (1) cellular and molecular pathways that stimulate repair after musculoskeletal injury, (2) biomechanical and biochemical cues in the microenvironment that influence repair, and (3) biomaterial-mediated tissue engineering approaches to promote the repair response.

Education: V.M.D. | University of Pennsylvania | 2017 Ph.D. in Bioengineering | University of Pennsylvania | 2016 B.S.E. in Biomedical Engineering, Duke University | 2009

Research/Work Experience: 2017–Present | Postdoctoral Research Fellow | Washington University in St. Louis | Advisor: Farshid Guilak, Ph.D. 2010–2016 | Graduate Research Assistant | University of Pennsylvania | Advisor: Robert Mauck, Ph.D. 2006–2009 | Undergraduate Research Assistant | Duke University | Advisor: George Truskey, Ph.D. 2003–2007 | Summer Research Fellow | Stony Brook University | Advisor: Miriam Rafailovich, Ph.D.

Selected Publications: • Qu F, Guilak F, Mauck RL. Cell Migration: Implications for Repair and Regeneration in Joint Disease. Nat Rev Rheumatol. 2019;15(3):167-179. • Qu F*, Holloway JL*, Esterhai JL, Burdick JA, Mauck RL. Programmed Biomolecule Delivery to Enable and Direct Cell Migration for Connective Tissue Repair. Nat Commun. 2017; 8(1):1780. *Co-first authors. • Qu F, Li Q, Wang X, Cao X, Zgonis MH, Esterhai JL, Shenoy VB, Han L, Mauck RL. Tissue Maturation State and Matrix Microstructure Regulate Interstitial Cell Migration in Dense Connective Tissues. Sci Rep. 2018; 8(1):3295. • Qu F, Pintauro MP, Haughan JE, Henning EA, Esterhai JL, Schaer TP, Mauck RL, Fisher MB. Repair of Dense Connective Tissues via Biomaterial-Mediated Reprogramming of the Wound Interface. Biomaterials. 2015; 39:85-94. • Qu F, Lin JG, Esterhai JL, Fisher MB, Mauck RL. Biomaterial-Mediated Delivery of Degradative Enzymes to Improve Meniscus Integration and Repair. Acta Biomater. 2013; 9(5):6393-402. • Qu F, Stoeckl BD, Gebhard PM, Hullfish TJ, Baxter JR, Mauck RL. A Wearable Magnet-Based System to Assess Joint Kinematics in Humans and Large Animals. Ann Biomed Eng. 2018; 46(12):2069-2078.

Awards/Honors: 2019–Present | NIH Ruth L. Kirschstein NRSA Postdoctoral Fellowship (F32) 2019 | New Investigator Recognition Award | Orthopaedic Research Society 2018–2019 | NIH T32 Postdoctoral Fellowship (Skeletal Disorders Training Program) 2015 | 1st Place, Ph.D. Competition | Summer Biomechanics, Bioengineering, and Biotransport Conference 2013–2015 | NIH T32 Predoctoral Fellowship (Training in Musculoskeletal Research) 2009–2012 | NIH T32 Predoctoral Fellowship (Medical Scientist Training Program) 2005–2009 | Angier B. Duke Memorial Scholarship (Full Merit Scholarship) | Duke University SATHISH RAMAKRISHNAN, PhD Nanobiology, Yale University, 100 West Campus Drive, Orange, CT 06477, [email protected]

Research Overview: My research program is primarily focused on neuroengineering – the design of biomaterials and technologies to help understand neurotransmission and to treat patients with neurological disorders. By interfacing neuroscience and engineering, I developed microarray and electro-optical technologies to mimic the brain pre-synaptic environment. I have characterized these technologies to interrogate the kinetics and key regulators involved in speed modulation of neurotransmitter and hormone release. These platforms can be applied to study memory formation and ultra-fast neuronal communication which will ultimately fuel research and improve treatments of neurological memory disorders; such as Alzheimer’s and Parkinson. During my PhD, I developed hybrid silicon photodetectors, point of care diagnostics, organic ultracapacitor for energy storage in collaboration with Storedot, Israel, and extensively studied the interface between biomolecules and inorganic materials. I developed several software packages for Raman's microscopy, super resolution microscopy, and single vesicle data analysis. My independent lab will focus on the development of biohybrid materials and technologies by utilizing my interdisciplinary skills in microfluidics, engineering, neuroscience, physics, and computer science to create real time voltage imaging probes to track changes in action potential, artificial neurons and synapses, and understanding of neural networks.

Education: Ph.D, in Physics, 2014, CNRS, University Montpellier, France M.S, in Applied Physics, 2009, Ecole Normale Superieure, France M.S, in Physics, 2008, University of Cambridge, United Kingdom B.Tech. in Biotechnology, 2006, Anna University, India

Research/Work Experience: 2018-Present: Associate Research Scientist, Yale University; Advisor: Prof. James Rothman (Nobel laureate) 2015-2018: Postdoctoral Associate, Yale University; Advisor: Prof. James Rothman 2011-2014: Graduate Research Assistant, CNRS, Montpellier, France; Advisor: Prof. Csilla Gergely 2009-2011, Research Support Assistant 2006-2007, Industrial Automation Programmer, SIEMENS, Bangalore, India

Selected Publications: Ramakrishnan Sk, et.al. FEBS Letters, 2019, 593: 154-162. Heo P, Ramakrishnan SK, et.al. Small, 2019, 1900725 Ramakrishnan SK, et.al. Langmuir, 2018, 34 (20), p 5849–5859 Coleman J, Ramakrishnan SK, et.al, Cell Reports, 2018, 22 (3), p820-831. Ramakrishnan SK, et.al. Sensors and Actuators B: Chemical, 2018, 272, p 211. Ramakrishnan SK, et.al. Langmuir 2016,32 (28), 7250–8, 2016

Awards/Honors: Submitted NIH Pathway to Independence Award (Parent K99/R00), Impact Score= 30, Oct 2018 Faculty Position at Tel Aviv University, Israel (Gratefully Declined) 2014 EGIDE Travel Grant for Hungary (2000 Euros, two times) 2012 & 2014 Award for most innovative idea, WUT, Wroclaw, Poland 2008 European Union Scholarship, (42,000 Euros) 2007 Academic Excellence award in Engineering, Anna University (two times) 2003 & 2004 National talent search award (62,000 INR) 2001 RITU RAMAN, PhD Koch Institute for Integrative Cancer Research, MIT, 500 Main Street, MIT 76-679, Cambridge, MA, 02142 - [email protected]

Research Overview: I am fascinated by the dynamically adaptive nature of biological materials, since their ability to sense, process, and respond to environmental signals in real-time enables high-level functional behaviors such as self-assembly, self- maintenance, and self-repair. I believe these capabilities must be integrated into engineered devices that interface with the human body, such as implants that sense and respond to individualized needs in real-time, to improve the standard of clinical care. My research is centered in “biohybrid design”, or using bio-integrated and bio-mimetic materials as functional components in engineered machines (i.e. muscle-actuated robots, light-degradable stents, heat-triggerable infusion pumps, etc.). I aim to establish a lab tackling pressing medical needs with biohybrid implants that dynamically monitor and therapeutically modulate the body to improve human health and quality-of-life.

Education: B.S., Mechanical Engineering, minor in Biomedical Engineering, magna cum laude; Cornell University (2008-2012) M.S., Mechanical Engineering; University of Illinois at Urbana-Champaign (2012-2013) Ph.D., Mechanical Engineering; University of Illinois at Urbana-Champaign (2013-2016)

Research/Work Experience: Postdoctoral Fellow, MIT (2017-Present) || Graduate Research Fellow, UIUC (2012-2016) Visiting Research Scholar, National University of Singapore (2014) || Undergraduate Researcher, Cornell University (2010-2012) Kessler Entrepreneurial Fellow, Rheonix, Inc. (2011) || Sub-Team Leader, AguaClara LLC. (2010)

Selected Publications: Full List: RituRaman.com/Publications [1] R.R.* (2019). Science, 363(6431). [2] R.R.* & R.L. (2019). Advanced Materials, 1901969. [3] L.G*, R.R.*, C.C., M.C.F., G.P.D., P.H., M.O.P & R.B. (2018). Tissue Engineering: Part A. *co-first author [4] R.R.*, C.C. & R.B. (2017). Nature Protocols, 12(3), pp.519-533. *Front Cover [5] R.R.*, L.G., Y.S., C.C., M.G., A.P., H.D., H.K., P.P. & R.B (2017). Advanced Healthcare Materials, 6(12).*Front Cover [6] R.R.* & R.B. (2017). Advanced Healthcare Materials, 6(20), 1700496. [7] L.R., B.R., A.W.F., R.R.*, K.K.P., R.B., S.M., P.D. & A.M. (2017). Science Robotics, 2, eaaq0495. [8] R.R.*, C.C., S.G.M.U., R.J.P., P.S., R.D.K. & R.B (2016). PNAS, 113(3), pp.3497-3502. [9] R.R.*, M.M., P.P., R.B. & L.D. (2016). Journal of Biological Engineering, 10(1), pp.10-18. [10] R.R.*, N.E.C., S.S., M.M., E.Q., H.K. & R.B. (2016). Biomedical Microdevices, 18(3), pp.1-7. *co-first author [11] R.R., B.B., M.M., A.S., M.K.L., G.P., H.K. & R.B. (2015). Advanced Healthcare Materials, 5(5). *Back Cover [12] J.A.S.N., R.R.*, V.C., M.G.R., M.S.B.R., J.J.V., R.L.D., R.B., P.T.H. & L.G. (2015). Biotech & Bioeng, 112(4), pp777-787. [13] C.C.*, R.R.*, V.C., B.J.W., M.T., P.B., M.S.S., H.H.A., M.T.A.S. & R.B. (2014). PNAS, 111(28), pp.10125. *co- first author.

Awards/Honors: Full List: RituRaman.com/Awards NASEM Ford Foundation Postdoctoral Fellow (2019) Sartorius & Science Prize for Regenerative Medicine & Cell Therapy (2019) MIT Technology Review 35 Innovators Under 35 (2019) AAAS IF/THEN Ambassador (2019) Koch Institute Postdoctoral Fellow (2019) Nature Research + Estée Lauder Inspiring Science Award Shortlist (2018) Forbes 30 Under 30: Science (2018) AAAS L’Oréal USA for Women in Science Fellow (2017) NSF EBICS Postdoctoral Fellow (2017) Koch Institute Marble Center Convergence Scholar (2017) MIT IMPACT Fellow (2017) BMES CMBE Student Fellow (2016) Baxter Young Investigator Award (2015) Illinois Innovation $15k Prize (2015) National Science Foundation Graduate Research Fellow (2014) SIYUAN RAO, PhD Postdoctoral Fellow in Bioelectronics Group, Massachusetts Institute of Technology, 77 Mass. Ave., Rm. 36-713, Cambridge, MA 02139 . [email protected] (857) 269-5168

Research Overview My overall career goal is to develop advanced material tools to facilitate the study of the biological system. In my postdoctoral career at MIT, together with my supervisors and colleagues, I have been developing remotely controlled magnetic material tools to enable temporally precise modulation of specific neural circuits underlying motivation and social interactions. I have applied custom-developed magnetic field apparatuses to wirelessly deliver magnetothermal stimuli into deep brain structures in freely moving, untethered mice. To achieve cell-type and circuit specificity, I have developed a rapid (<20 sec latency) magnetically controlled chemical payload release approach. By selecting specific ligand-receptor pairs as a means to chemically modulate neural activity, for the first time, I realized the repeated control of multiple behaviors in freely moving mice via remote magnetic stimulation. My graduate school research has been focused on proteorhodopsin (pR) based bio-hybrid photoelectric energy conversion artificial systems. pR-integrated microdevices, such as photocurrent generators, bio-capacitors and pH sensors, were designed and constructed into electronics application. These findings provide new thoughts to use biophysical insights to assist material engineering, as well as to utilize artificial approaches to modulate biological processes. In my future independent career, I plan to utilize the previous training and experience to explore the multidisciplinary fields of material-neural interfaces for targeted neural modulation and chronic neural activity recording.

Professional Preparation • Massachusetts Institute of Technology Cambridge, MA Postdoctoral Fellow in Bioelectronics and Neuroengineering 2016-present • Beihang University Beijing, China Ph.D. in Material Physics and Chemistry 2010-2015 • Monash University Melbourne, Australia Exchange Graduate Student in NanoBionics Group 07/2014-09/2014 • Beihang University Beijing, China Bachelor of Engineering 2006-2010

Selected Publications 1) Rao S, Chen R, LaRocca A, Christiansen M, Senko A, Shi C, Chiang P, Varnavides G, Xue J, Yang Z, Park S, Ding R, Moon J, Feng G, Anikeeva P. Remotely controlled chemomagnetic modulation of targeted neural circuits, Nat. Nanotechnology 2019; doi: 10.1038/s41565-019-0521-z. 2) Rao S, Si KJ, Yap LW, Xiang Y, Cheng W. Free-Standing Bilayered Nanoparticle Superlattice Nanosheets with Asymmetric Ionic Transport Behaviors. ACS Nano. 2015; 9(11):11218-24. 3) Rao S, Guo Z, Liang D, Chen D, Li Y, Xiang Y. 3D proton transfer augments bio-photocurrent generation. Adv Mater. 2015; 27(16):2668-73. 4) Rao S, Xiu R, Si J, Lu S, Yang M, Xiang Y. In situ synthesis of nanocomposite membranes: comprehensive improvement strategy for direct methanol fuel cells. ChemSusChem. 2014; 7(3):822-8. 5) Rao S, Lu S, Guo Z, Li Y, Chen D, Xiang Y. A light-powered bio-capacitor with nanochannel modulation. Adv Mater. 2014; 26(33):5846-50. 6) Rao S, Guo Z, Liang D, Chen D, Wei Y, Xiang Y. A proteorhodopsin-based biohybrid light-powering pH sensor. Phys Chem Chem Phys. 2013; 15(38):15821-4.

Awards and Honors 2019 NIH Pathway to Independence Award (K99/R00, NIMH, K99MH120279) 2016 Simons Postdoctoral Fellowship, Simons Foundation to the Simons Center for the Social Brain at MIT 2015 Excellent Graduate Student, Beihang University 2014 National Scholarship, Ministry of Education of the People's Republic of China 2010 Outstanding Undergraduate of Beijing, Beijing Municipal Administration Committee 2010 Excellent Undergraduate Student, Beihang University JIAN REN, PhD Wellman Center for Photomedicine, Harvard Medical School, 40 Blossom Street, BAR 822, Boston, MA, 02114 [email protected]

Research Overview: My research lies in the field of biophotonics, specializing in biomedical imaging and neurophotonics. I aim to understand light interactions with bio-systems and utilize these interactions to create transformative tools. For basic research, my focus is to probe and analyze large-scale complex biological systems, such as intact brains, in a systematic and scalable manner. I conceived and developed Clearing Assisted Scattering Tomography (CAST), a 3D high-throughput imaging procedure, enabling optical investigation in biological systems without the need of physical tissue sectioning. Distinct from traditional bio-optics works, I not only develop imaging instrumentation but also innovate tissue processing and build computational tools. Deeply fusing optical, biochemical, and computational approaches, this work would provide potential technical solutions to help answer many open questions in research areas like connectomics and Alzheimer's. For translational research, I strive to develop enabling medical tools empowered by new photonic devices and to integrate complementary modalities for diverse clinical applications. I have built various preclinical imaging probes and systems, which have shown early promise in providing intraoperative guidance for ophthalmology and orthopedics. I helped found oProbe LLC to commercialize these technologies and served as its Chief Technology Officer in 2013. In my current position, I have been playing a key role in the first clinical studies on coronary artery disease using polarization-sensitive imaging.

Education: Ph.D. Electrical Engineering, California Institute of Technology, Pasadena, CA, 2013 M.S. Electrical Engineering, University of California, San Diego, CA, 2007 B.S. & M.E. , Tsinghua University, Beijing, China, 2004

Research/Work Experience: 2013 – current, Research Fellow, Massachusetts General Hospital, Harvard Medical School, Boston, MA 2010 – 2013, Consultant & Chief Technology Officer, oProbe LLC, Pasadena, CA 2008 – 2012, Research Assistant, California Institute of Technology, Pasadena, CA 2010, Consultant, Visualyze Technologies Inc., Los Angeles, CA 2007, Firmware Engineer, Qualcomm Inc., San Diego, CA 2004 – 2006, Research Assistant, University of California, San Diego, CA 2001 – 2004, Research Assistant, Tsinghua University-Bell Labs Joint Laboratory, Beijing, China

Selected Publications: · Jian Ren, H. Choi, K. Chung and B. E. Bouma, ‘Label-free volumetric optical imaging of intact murine brains’, Scientific Reports, vol. 7, 46306; doi: 10.1038/srep46306, April 2017 · Jian Ren, ‘Dispersion analysis and measurement of potassium tantalate niobate crystals by broadband optical interferometers’, Applied Optics, vol. 56, no. 2, pp. 234-238, January 2017 · Jian Ren, X. Cui, L. M. Lee and C. Yang, ‘Quantitative surface normal measurement by a wavefront camera’, Optics Letters, vol. 37, no. 2, pp. 199-201, January 2012 · Jian Ren, H. K. Gille, J. Wu and C. Yang, ‘Ex vivo optical coherence tomography imaging of collector channels with a scanning endoscopic probe’, Investigative Ophthalmology & Visual Science, vol. 52, no. 7, pp. 3921-3925, June 2011 · Jian Ren, J. Wu, E. J. McDowell and C. Yang, ‘Manual-scanning optical coherence tomography probe based on position tracking’, Optics Letters, vol. 34, no. 21, pp. 3400-3402, November 2009

Awards/Honors: · Pathway to Independence Award (K99/R00), National Institute on Aging, National Institutes of Health, 2019–2024 · Best Paper Award, Gordon Research Conference: Lasers in Medicine and Biology, West Dover, VT, 2016 · Selected to Future Faculty ADVANCE Program, National Science Foundation, 2012 · OCT News Student Travel Grant Award, 2012 · Powell Fellowship, California Institute of Technology, 2008 · Guanghua Fellowship, Tsinghua University, 2002 · Scholarship for Excellency in Science & Technology Competition, First Class Award, Tsinghua University, 2000 · First prize of the 15th University Student Physics Contest of Beijing, 1999 RACHEL RILEY, PhD Bioengineering, University of Pennsylvania, 210 South 33rd Street, Suite 240, Skirkanich Hall, Philadelphia, PA, 19104 [email protected]

Research Overview: My research interests lie at the interface of disease management, nanotechnology, and engineering to develop novel drug delivery technologies to treat understudied topics related to women’s health. During my graduate studies in Dr. Emily Day’s lab at the University of Delaware, I developed gold-based nanoparticles for precise control over treating triple- negative breast cancer (TNBC) using gene therapy and light activation. Through this research, I showed that attaching antibodies or siRNA to nanoparticles enables active targeting of TNBC or on-demand gene regulation, respectively. By exploiting the unique physicochemical characteristics of these gold nanoparticles, I demonstrated how single nanoparticle platforms can be used to treat cancer via multiple mechanisms (i.e. gene regulation and light-mediated thermal therapy) simultaneously. My postdoctoral research utilizes lipid and polymer-based nanoparticles as platforms to deliver therapeutic nucleic acids such as siRNA or mRNA to treat fetal diseases. In this research, I engineered new lipid-based materials for nucleic acid delivery to fetuses in utero that can be used to enable gene regulation to treat diseases early in gestation. My overarching goal is to utilize these research experiences to study and treat issues pertaining to women’s health through two distinct research focuses: (1) development of drug delivery technologies to enable mRNA vaccination and siRNAmediated gene silencing to treat solid tumors such as breast and ovarian cancer, and (2) engineer biomaterial platforms to treat prenatal diseases and prevent preterm birth. Thus, my independent research program will take an interdisciplinary approach combining engineering, nanotechnology, and gene therapy to develop novel drug delivery platforms to study and treat diseases and disorders related to women’s health.

Education: Ph.D., Biomedical Engineering, 2018, University of Delaware BS, 2012, Rowan University

Research/Work Experience: NIH T32 Postdoctoral Fellow, University of Pennsylvania, 2018-Present, Advisor: Dr. Michael Mitchell Ph.D. Student, University of Delaware, 2013-2018, Advisor: Dr. Emily Day Engineer I and Intern, Pepco Holdings, Inc., 2011-2013

Selected Publications: 1. Riley RS*, Kashyap M*, Billingsley MM*, Peranteau W, Mitchell MJ. Discovery of Lipid Nanoparticles for In Utero Delivery of Therapeutic Nucleic Acids. In Preparation. *contributed equally. 2. Riley RS, June C, Langer R, Mitchell M. Delivery Technologies for Cancer Immunotherapies: Challenges and Opportunities (2018). Nature Reviews Drug Discovery. 18(3), pp 175-196. 3. Riley RS*, O’Sullivan RK*, Potocny AM, Rosenthal J, Day ES. Evaluating Nanoshells and a Potent Biladiene Photosensitizer for Dual Photothermal and Photodynamic Therapy of Triple Negative Breast Cancer Cells (2018). Nanomaterials. 8(9), pp 658. *contributed equally. 4. Potocny AM*, Riley RS*, O’Sullivan RK, Day ES, Rosenthal J. Photochemotherapeutic Properties of a Linear Tetrapyrrole Palladium(II) Complex displaying an Exceptionally High Phototoxicity Index (2018). Inorganic Chemistry. 57(17), pp 10608- 10615. *contributed equally. 5. Riley RS, Dang M, Billingsley MM, Abraham B, Gundlach, L, Day ES (2018). Evaluating the Mechanisms of Light- Triggered siRNA Release from Nanoshells for Temporal Control Over Gene Regulation. Nano Letters. DOI: 10.1021/acs.nanlett.8b00681. 6. Riley RS, Day ES (2017). Frizzled7 Antibody-Functionalized Nanoshells Enable Multivalent Binding for Enhanced Wnt Signaling Inhibition in Triple Negative Breast Cancer Cells. Small. 13, 1700544.

Awards/Honors: Best Poster Travel Award, Gordon Research Conference in Cancer Nanotechnology, 2019 Best Doctoral Thesis Award, University of Delaware, 2019 NIH T32 Fellow in Multidisciplinary Cardiovascular Training Grant, University of Pennsylvania, 2018 Biomedical Engineering Distinguished Scholar Award, University of Delaware, 2018 American Association of University Women (AAUW), Dissertation Fellow, 2017 University of Delaware Graduate Fellowship, 2016 NSF Graduate Research Fellowship Program Honorable Mention, 2015 DEKEL ROSENFELD, PhD Research laboratory of engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Camnridge, MA, 02139 [email protected]

Research Overview: In my PhD I studied the involvement of mechanical forces in: 1)embryonic stem cells differentiation in three-dimensional environment of polymeric scaffolds 2) organization of blood vessels network in vascularized engineered tissues. My current research is in the field of Bioelectronic medicine and is focused on using magnetic nanoparticles and alternating magnetic fields to stimulate heat sensitive ion channels to evoke action potentials in electroactive cells. Specifically, we developed a method to control remotely the stress hormone release from adrenal gland using the approach of magneto-thermal stimulation, relevant for the regulation of stress hormone in stress-related diseases. I am applying the magnetothermal approach both for peripheral organ modulation and for controlling neuronal growth within three-dimensional matrices. My research interests include: Bioelectronic medicine, neuro-engineering, magnetic nanoparticles, adrenal hormone release, biomaterials,o rganization and functionality of engineered tissues and cells under external stimulation.

Education: 2014: Doctor of Philosophy, Department of Biomedical Engineering, Technion, Israel. 2009:Master of Science, Department of Biomedical Engineering, Technion, Israel. 2007:Bachelor of Science, Department of Biomedical Engineering, Technion, Israel

Research/Work Experience: Post-doctoral fellow, Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, MA, USA. Supervisor: Prof. Polina Anikeeva Post-doctoral fellow, Department of Biomedical Engineering, Technion, Israel Supervisor: Prof. Shulamit Levenberg

Selected Publications: 1)Dekel Dado and Shulamit Levenberg. Cell–scaffold mechanical interplay within engineered tissue. Seminars in Cell and Developmental Biology.2009. 2)Dekel Dado-Rosenfeld, Itai Tzchori, Amir Fine, Limor Chen-konak and Shulamit Levenberg. Tensile forces applied on a cellembedded 3D scaffold can direct early differentiation of embryonic stem cells toward the . Tissue engineering Part A. 2014 3)Yulia Shandalov, Dana Egozi, Jacob Koffler, Dekel Dado-Rosenfeld, David Ben-Shimol, Alina Freiman, Erez Shor, Aviva Kabala, and Shulamit Levenberg. An engineered muscle flap for reconstruction of large soft tissue defects. Proc Natl Acad Sci U S A. 2014 4)Dekel Rosenfeld, Shira Landau, Yulia Shandalov, Noa Raindel, Erez Shor, Yaron Blinder, Herman Vandenburgh, David Mooney and Shulamit Levenberg. Morphogenesis of 3D vascular network is regulated by tensile forces. Proc Natl Acad Sci U S A.2016. 5)Jonathan Avesar, Dekel Rosenfeld, Marianna Truman-Rosentsvit, Tom Ben Arye, Yuval Geffen, Moran Bercovici, and Shulamit Levenberg. Rapid phenotypic antimicrobial susceptibility testing using nanoliter arrays. Proc Natl Acad Sci U S A, 2017 6)Dekel Rosenfeld, Alexander W. Senko, Junsang Moon, Isabel Yick, Georgios Varnavides, , Danijela Gregureć, Florian Koehler, Po-Han Chiang, Michael G. Christiansen, Lisa Y. Maeng, Alik S. Widge and Polina Anikeeva. Remote Magnetothermal Control of Adrenal Hormones. Submitted

Awards/Honors: 1)NIH BRAIN Initiative Trainee award, 2018 2)Israel VATAT fellowship for Women in science, 2018 3)MIT-Technion Fellowship for post-doctoral research, MIT, 2016. 4)Israel VATAT fellowship for Women in science, 2017 5)Ed Sattel scholarship for excellent PhD students, 2009-2014. 6)Excellence in Teaching Award, Technion, Israel, 2012. 7)Excellence in Teaching Award, Technion, Israel, 2013 8)Katzir fellowship for short term internship abroad, Boston, MA, 2012 9)Diploma with honors (cum laude), Biomedical engineering department, Technion, Israel, 2007. 10)Oral presentation award of the Israel Stem Cell Society, ISCS conference, Israel, 2014 ALEXANDRA RUTZ, PhD Electrical Engineering, University of Cambridge, 9 JJ Thomson Ave, Cambridge, Cambridgeshire, CB3 0FA, United Kingdom . [email protected]

Research Overview: Bioelectronics are devices that mediate the flow of information between living systems and our technology. These devices have become life-changing therapeutic and diagnostic technologies in healthcare as well as pillar instruments for studying and monitoring biological phenomena. Over the next decades, the impact of bioelectronics is projected to greatly expand; however, poor tissue electronic Interfacing limits application and efficacy of devices. In order to improve biointegration and enable new applications, I propose research directions that lie at the intersection of bioelectronics, biomaterials, tissue engineering, and 3D printing. My research vision is to merge electronics and biology by fabricating them together into a single device. I will pioneer electronic tissues: engineered tissues containing a network of embedded electronics. Towards this goal, I will advance the structure of electronic circuitry from planar and solid configurations to those that are three-dimensional and porous, thus building electronic device-scaffold hybrids for tissue engineering. I will also progress inert bioelectronic materials into those that promote specific and beneficial biological processes by incorporating components based on natural extracellular matrix. Finally, I will address uncontrolled and nonspecific tissue-electronic interfacing by developing additive manufacturing materials and techniques. Simultaneous fabrication of tissue and electronics will allow me to precisely connect cells with electronic elements. With these advances, the long-term goals of my lab include designing next-generation implantable devices, providing platforms for continuous, on-line monitoring of tissues-on chips, and developing electronic, regenerative engineering therapeutics.

Education: PhD | Northwestern University, Biomedical Engineering, 2016 BS | University of Illinois Urbana-Champaign, Chemistry & Molecular and Cellular Biology, 2011

Research/Work Experience: Postdoctoral | 2016-Present | University of Cambridge, Advisor: George G. Malliaras (Bioelectronics) Slippery coatings for reducing insertion trauma of implantable neural probes (Poster #523) Graduate | 2011-2016 | Northwestern University, Advisor: Ramille N. Shah (Biomaterials, Tissue Engineering, 3D Bioprinting) Hydrogel ink and bioink development for 3D printing tissues, including a functional bioprosthetic ovary Undergraduate | 2008-2011 | UIUC, Advisor: Steven C. Zimmerman (Organic Chemistry)

Selected Publications: 1. Zeglio E, Rutz AL, Winkler TE, Malliaras GG, Herland A. “Conjugated polymers for assessing and controlling biological functions,” Advanced Materials 2019, 1806712. 2. Pas J, Rutz AL, Quilichini P, Slézia A, Ghestem A, Kaszas A, Donahue M, Curto V, O’Connor RP, Bernard C, Williamson A, Malliaras GG. “A bilayered PVA/PLGA-bioresorbable shuttle to improve the implantation of flexible neural probes,” Journal of Neural Engineering 2018 15 (6), 065001. 3. Rutz AL, Lewis PL, Shah RN. “Toward next-generation bioinks: tuning material properties pre- and post-printing to optimize cell viability,” MRS Bulletin 2017, 42. 4. Rutz AL* and Laronda MM*, Whelan KA, Roth EW, Woodruff TK, Shah RN. “A bioprosthetic ovary created using 3D printed microporous scaffolds restores ovarian function in sterilized mice,” Nature Communications 2017, 8. [Widely covered by media outlets, Altmetric Top 100 articles of 2017] *, co-first authors 5. Rutz AL, Hyland KE, Jakus AE, Burghardt WR, Shah RN. “A multi-material bioink method for 3D printing tunable and cellcompatible hydrogels,” Advanced Materials 2015, 27 (9) 1607-1614.

Awards/Honors: 2018-Present| Marie Skłodowska-Curie Actions Individual Fellowship (€183,000 EUR) 2018| Whitaker International Program’s Concluding Initiative Grant ($50,000 USD) 2018| University of Cambridge Engineering for Clinical Practices Grant (£10,000 GBP) 2017| MilliporeSigma Life Science Award in 3D Printing, First Place ($10,000 USD) 2016-18| Whitaker International Post-doctoral Scholarship ($100,000 USD) 2015| Baxter International Inc. Young Investigator Award 2015| STAR Award, Society for Biomaterials 2013-16| NSF Graduate Research Fellowship ($150,000 USD) EIJI SAITO, PhD University of Michigan, 1600 HURON PKWAY, Ann Arbor, MI, 48109-2099 [email protected]

Research Overview: My research laboratory will focus on development of biomaterials for immunomodulation and tissue regeneration. Trauma or disease (e.g. autoimmunity) have associated inflammation, with immune cells infiltrating to damage the tissue. Therapy must overcome both the immune environment and also regenerate the damaged tissue. I aim to develop immunomodulatory “drug- free” nanoparticles, without any active ingredient, to treat inflammation and injury. Furthermore, I will develop antigen-specific nanoparticle therapy for autoimmune disease. Subsequently, my laboratory will develop bioactive scaffolds that modulate immune cells, especially macrophages, to promote tissue regeneration. Importantly, the nanotherapies and biomaterial scaffolds provide tools to modulate immune responses and tissue regeneration, which my research will exploit to investigate the mechanisms relating immunomodulation and tissue regeneration. Nanoparticles for immunomodulation: In Prof. Lonnie Shea’s lab, I have been developing biodegradable nanoparticles for treatment of innate inflammation and autoimmune disease using mouse models, including spinal cord injury (SCI) and experimental autoimmune encephalomyelitis (EAE). For mitigating innate inflammation, I have developed “drug-free” nanoparticles to target inflammatory monocytes and neutrophils in the vasculature and redirect them to the spleen or liver rather than the primary site of inflammation, brain and spinal cord. I have also investigated various biodegradable nanoparticles loaded with disease-specific antigen and various administration routes of the particles to treat EAE. The particles reduced the expression of the costimulatory molecules on APCs, which led to deactivation of CD4+ T-cells. I am currently developing particles to reprogram macrophages for tissue regeneration. 3D printed porous scaffolds for tissue regeneration: In Prof. Scott Hollister’s lab, the focus of my work was the development of biodegradable porous scaffolds for orthopaedic applications using computational design and 3D printing. I investigated the effect of scaffold materials, architecture, and surface coating on tissue formation and scaffold degradation to fill bone defects. My research showed that scaffold architectures determined in vivo degradation of scaffolds. I also demonstrated that scaffold materials influenced in vivo tissue regeneration on and into porous scaffolds. My oral presentation at the BMES Annual Meeting is scheduled for 2:30pm on 10/17/2019

Education: PhD, University of Michigan-Ann Arbor, Biomedical Engineering, December 2011, Advisor: Scott J. Hollister, PhD BS, Kanazawa University, Japan, Mechanical Engineering

Research/Work Experience: • University of Michigan, Ann Arbor, MI, 2014-Present, Postdoctoral Research Fellow, Advisor: Lonnie D. Shea, PhD • Northwestern University, Chicago, IL, 2013-2014, Postdoctoral Research Fellow, Advisor: Lonnie D. Shea, PhD • University of Michigan, Ann Arbor, MI, 2012-2013, Postdoctoral Researcher, Advisor: Scott J. Hollister, PhD • University of Michigan, Ann Arbor, MI, 2003-2011, Graduate Student Research Assistant, Advisor: Scott J. Hollister, PhD

Selected Publications: 1. Eiji Saito, Robert Kuo, Kevin R. Kramer, Nishant Gohel, David A. Giles, Bethany B. Moore, Stephen D. Miller, Lonnie D. Shea,“Design of Biodegradable Nanoparticles to Modulate Phenotypes of Antigen Presenting Cells for Antigen-Specific Treatment of Autoimmune Disease” Biomaterials 2019 (Accepted, Article in Press) 2. Eiji Saito, Robert Kuo, Ryan M. Person, Nishant Gohel, Brandon Cheung, Nikolas J.C. King, Stephen D. Miller, Lonnie D. Shea, “Designing Drug-free Biodegradable Nanoparticles to Modulate Inflammatory Monocytes and Neutrophils for Ameliorating Inflammation” Journal of Controlled Release 2019 Apr 28; 300:185-196 3. Eiji Saito, Darilis Suárez-Gonzále, William L. Murphy, Scott J. Hollister, “Biomineral Coating Increase Bone Formation by Ex-Vivo BMP-7 Gene Therapy in Rapid Prototyped Poly (L-lactic acid) (PLLA) and Poly (ε-caprolactone) (PCL) Porous Scaffolds” Advanced Healthcare Materials 2015 Mar 11; 4(4): 621-32 4. Eiji Saito, Yifie Liu, Francesco Migneco, Scott J. Hollister, “Strut Size and Surface Area Effects on Long Term In Vivo Degradation in Computer Designed Poly (L-lactic acid) 3D Porous Scaffolds” Acta Biomaterialia 2012;8:2568-2577 5. Eiji Saito, Elly E. Liao, Wei-Wen Hu, Paul H. Krebsbach, Scott J. Hollister, “Effects of designed PLLA and 50:50PLGA scaffold architectures on bone formation in vivo”, J Tissue Eng Regen Med 2013 7(2): 99-111

Awards/Honors: University of Michigan Rackham Graduate Student Research Grant (2011) University of Michigan Rackham Conference Travel Grant (Fall:2008, 2009, 2011, Winter: 2011) REBECCA D. SANDLIN, PhD Center for Engineering in Medicine, Harvard Medical School, Massachusetts General Hospital, 114 16th St., Rm 1239, Charlestown, MA, 02129 . [email protected] Research Overview: I am an Instructor in Surgery at Harvard Medical School and Massachusetts General Hospital. My research program is focused on the application of engineering technologies and concepts to solve critical problems in global health. My graduate training was in chemistry with an emphasis on antimalarial drug discovery and malaria parasite biochemistry, while my postdoctoral training was in bioengineering (PI: Mehmet Toner) and focused on cryobiology and the development of biostabilization methods for microfluidic processing of cells. As a junior faculty member, I have merged my graduate and postdoctoral training to establish a global health program within the Center for Engineering in Medicine. Specifically, my lab is focused on: (1) Development of point-of-care diagnostics for asymptomatic malaria (R21, PI: Sandlin); (2) Development of biostabilization methods for global health applications (R21, PI: Sandlin/Usta; CDC subcontract, PI: Sandlin; Gates Foundation, PI: Tzipori) and (3) Elucidation of host-pathogen interactions of biocrystalline malaria metabolites and the immune system.

Education: Ph.D. Chemistry, 2013; Vanderbilt University B.S. Chemistry and Mathematics, 2007; Western Kentucky University

Research/Work Experience: • Instructor in Surgery (Dec.2016-present) Center for Engineering in Medicine, Harvard Medical School/Massachusetts General Hospital (HMS/MGH) • Postdoctoral Research Fellow (2013-2016) Center for Engineering in Medicine, (HMS/MGH) • Graduate Research Assistant (2008-2013) Department of Chemistry, Vanderbilt University • Teaching Assistant (2008-2010) Department of Chemistry, Vanderbilt University • Research Assistant (2005-2008) Department of Chemistry, Western Kentucky University

Selected Publications: 1. Jaskiewicz, J.J.*; Sandlin, R.D.*; Swei, A.A.; Widmer, G; Toner, M.; Tzipori,S. Cryopreservation of infectious Cryptosporidium oocysts. Nature Communications 2018:9:2883. *denotes co-first author 2. Sandlin, R.D.; Wong, K.H-K.; Tessier, S.N. et al. Ultra-fast vitrification of patient-derived circulating tumor cell lines. PLoS one 2018;13(2):e0192734. 3. Sandlin, R.D.*; Fong, K.Y.*; Stiebler, R.; et al. Detergent-Mediated Formation of beta-Hematin: Heme Crystallization Promoted by Detergents Implicates Nanostructure Formation for Use as a Biological Mimic. Crystal Growth & Design 2016; 16(5):2542-51. 4. Sandlin, R.D.; Fong, K.Y.; Wicht, K.J.; et al. Identification of beta-hematin inhibitors in a high-throughput screening effort reveals scaffolds with in vitro antimalarial activity. International Journal of Parasitology: Drugs and Drug Resistance. 2014; 4(3):316-25. 5. Sandlin, R.D.; Starling, M.P.; Williams, K.M. A bulky platinum triamine complex that reacts faster with guanosine 5'- monophosphate than with N-acetylmethionine. Journal of Inorganic Biochemistry 2010; 104(2):214-6.

Awards/Honors: • 2019-present, NIH/NIAID R21 Award (1R21AI137558-01A1) “Cryogenic purification of Plasmodium parasites from blood” (PI: Sandlin) • 2018-present, NIH/NIAID R21 Award (1R21AI142415-01) “High-Quality Long-Term Preservation of Sporozoites via Deep Supercooling” (PI: Sandlin/Usta) • 2018-present, CDC Subcontract ($376k in Direct Costs) “Mosquito Cryopreservation and Female Elimination” (PI: Sandlin) • Poster awards: 2014 (Gordon Conference); 2012 (Keystone Symposia); 2005 (Sigma Xi Student Research) • Oral presentation awards: 2007/2006 (Sigma Xi Student Research); 2006 (Kentucky Academy of Sciences) • Academic Honors: 2012 Travel Award (Keystone Symposia); 2007 (Outstanding Senior in Chemistry); 2006 (C.P. McNally Award in Chemistry); 2005 (Pi Mu Epsilon) • Teaching Fellow, Vanderbilt University, 2009/2010 Selected to teach problem solving companion course for general chemistry (~100 students/semester) BIPLAB SARKAR, PhD Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St, Newark, NJ, 07102 [email protected]

Research Overview: As a researcher in Biomedical Engineering, I am interested in developing peptide-based materials for tissue regeneration. These injectable materials can act as regenerative scaffolds for cells in vivo and tune the body’s response to injury and inflammation. Through supramolecular chemistry, the materials properties and biological functionalities of these hydrogels can be adjusted for use in specific tissues and organs, such as vitreous humor, dental pulp, and brain parenchyma. I have 8+years’ experience in the design of proteins, peptides, & antibodies, and their analysis through spectroscopy, microscopy, and chromatography. I am a co-Founder of a biotech startup targeted at treating neovascular ocular diseases (SAPHTx, Inc.). I was previously a research intern at MedImmune (AstraZeneca). I have trained and mentored 12 undergraduate students and 3 graduate students at Rice, Tufts, and NJIT. My research has been featured in press conferences at the American Chemical Society meetings (Fall 2018 and Fall 2019 ACS national meetings) and in popular press (such as ScienceDaily and FierceBiotech). I teach the undergraduate General Chemistry class at NJIT and write on behalf of the Materials Research Society at MRS Meeting Scene and Materials Connect blogs, focusing on self- assembly and biomaterials.

Education: •Rice University, 2015. PhD, Chemistry • Indian Institute of Technology, 2010. MSc, Chemistry

Research/Work Experience: • Postdoctoral Research Associate, 2017 - present New Jersey Institute of Technology; Advisor: Vivek A. Kumar, PhD • Co-Founder, 2018 – present SAPHTx, Inc. • Postdoctoral Scholar, 2015 – 2017 Tufts University; Advisor: David L. Kaplan, PhD • Intern, Summer 2015 MedImmune (AstraZeneca) • Graduate Research Assistant & Teaching Assistant, 2010 – 2015 Rice University; Advisor: Jeffrey D. Hartgerink, PhD

Selected Publications: * corresponding author; ‡ equal contribution; # mentored undergraduate researcher 1. Sarkar, B.; O’Leary, L. E. R.; Hartgerink, J. D.* J. Am. Chem. Soc. 2014, 136, 14417–14424. 2. Nguyen, P. K.; Gao, W.#; Patel, S.D.#; Siddiqui, Z.; Weiner, S.; Shimizu, E.; Sarkar, B.*; Kumar, V. A.* ACS Omega 2018, 3 (6), 5980–5987. 3. Sarkar, B.; Nguyen, P. K.; Gao, W.#; Dondapati, A.#; Siddiqui, Z.; Kumar, V. A.* Biomacromolecules 2018, 19 (9), 3597–3611. 4. Nguyen, P. K.‡; Sarkar, B.‡*; Siddiqui, Z.; McGowan, M; Iglesias-Montoro, P.#; Rachapudi, S.#; Kim, S.; Gao, W. #; Lee, E. J.; Kumar, V. A.* ACS Appl. Bio Mater. 2018, 1 (3), 865-870. 5. Sarkar, B.‡; Siddiqui, Z.‡; Nguyen, P. K.‡; Dube, N.; Fu, W.; Park, S.; Jaisinghani, S.#; Paul, R.#; Kozuch, S. D.; Deng, D.; Iglesias-Montoro, P.#; Li, M.; Sabatino, D.; Perlin, D. S.; Zhang, W.; Mondal, J.; Kumar, V.A.* ACS Biomater. Sci. Eng. 2019, DOI: 10.1021/acsbiomaterials.9b00967.

Awards/Honors: • National Science Foundation I-Corps Award (2019) • DST INSPIRE Award from the Government of India (2019) • MedImmune Summer Intern Award (for best performance in the program) (2015) • Robert A. Welch Foundation Pre-doctoral Fellowship (Rice University) (2013) • Evans Atwell-Welch Graduate Fellowship (Rice University) (2010)

BENJAMIN L SCHWARTZ, PhD School of Biological and Health , Arizona State University, Tempe, AZ 85287 [email protected]

Research: Future directions: I intend to maintain a laboratory program studying the interface of neurons and invasive and non-invasive electrodes, broadly divided into two categories: 1) fluorescent imaging of intracellular metabolites and 2) MR electrical impedance tomography. A third category I wish to pursue is expanding the use of MRI for indirectly measuring material behavior. I’ve studied now to obtain, non-invasively, quantitative measures of a material’s complex shear modulus with MRE and it’s electrical conductivity with MREIT. Hæmodynamics are critically important to brain function; so, I aim to extend MRI’s use to study fluid flow structures in major cerebral blood vessels. For whatever scientific infrastructure is lacking, I will write major research instrumentation grants, in addition to grants directly funding my work. All the projects I intend to do are interdisciplinary which broadens the field of grant awarding agencies I may ask for money. Implementing my research plans will employ existing laboratory resources while developing new analytic capabilities. Mine will be a vigorous program that contributes to our knowledge of the point where human meets machine. Work to Present: My work has advanced the understand of living tissue’s interaction with diagnostic devices on several fronts. My series elastic airway model is often cited as a demonstration of the heterogeneous nature of lung mechanics and why more sophisticated mathematic representations of such systems are needed. My doctorate establishes the modeling methods for MR elastography of complex geometries. My current work focuses on electromagnetic field theory and is expended to fully three dimensional models of conventional, though unexplored in this context, geometries. The end goal is then, by characterizing the electromagnetic fields neural tissue experiences during neuromodulation and neuroimaging, to create predictive, testable models of neural activation by those fieled.Such detailed knowledge will help us understand e.g. the biophysical processes in something as fundamental as an action potential and something more directly clinically germane like pathologic neurodegeneration.

Education: _ University of Illinois at Chicago, August 2014. PhD, Bioengineering _ University Vermont, October 2009. MS, Bioengineering _ Arizona State University, May 2006. BSE, Bioengineering

Experience: _ Resident Faculty, Engineering, Phoenix College, 2019–present _ Visiting Scholar, Neural Microsystems Laboratory; PI: Jit Muthuswamy, PhD, 2016–present _ Postdoctoral Scholar, Neuroelectricity Laboratory; PI: Rosalind Sadleir, PhD, 2014–2016 _ Research Fellow, Diagnostic NMR Systems Laboratory; Richard Magin, PhD, 2010–2014 _ Graduate Researcher, Vermont Lung Center; PI: Jason Bates, PhD, DSc, 2007–2009

Selected Publications: 1. Benjamin L. Schwartz, Ron C. Anafi, Minara Aliyeva, John A. Thompson-Figueroa, Gilman B. Allen, Lennart K. A. Lundblad, and Jason H. T. Bates. Effects of central airway shunting on the mechanical impedance of the mouse lung. Ann Biomed Eng 39(1):497–507, 2011. 2. Benjamin L. Schwartz, Yifei Liu, Thomas J. Royston, and Richard L. Magin. Axisymmetric diffraction of a cylindrical transverse wave by a viscoelastic spherical inclusion. J Sound Vib 364:222–233, 2016. 3. Benjamin L. Schwartz, Ziying Yin, and Richard L. Magin. Mechanical analysis of an axially symmetric cylindrical phantom with a spherical heterogeneity for MR elastography. Phys Med Biol 61(18):6821–6832, 2016. 4. Benjamin L. Schwartz, Ziying Yin, Temel K. Yas¸ar, Yifei Liu, Altaf Kahn, Allen Ye, Thomas J. Royston, and Richard L. Magin. Scattering and diffraction of elastodynamic waves in a concentric cylindrical phantom for MR elastography. IEEE Trans Biomed Eng 63(11):2308–2316, 2016. 5. Benjamin L. Schwartz, Munish Chauhan, and Rosalind J. Sadleir. Analytic modeling of conductively anisotropic neural tissue. J Appl Phys 124(6):064701, 2018. ISMAEL SEÁÑEZ, PhD Center for Neuroprosthetics and Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), Geneva, Switzerland . [email protected]

Research Overview: Every neuromotor disorder, like spinal cord injury (SCI) and stroke, is unique for each individual. Therefore, it is crucial to develop the next generation of personalized therapies that continuously adapt to individual residual function and potential for recovery. During my doctoral research, I developed a non-invasive body-machine interface (BoMI) for people with high-level SCI that adapted to individual residual shoulder movements and converted them into 2D signals to control a computer cursor, play videogames, and drive a wheelchair1,2. This BoMI provided severely impaired people with a powerful tool to control assistive devices3, and a device for rehabilitation that promoted the reorganization process of the brain and body4. During my postdoctoral work, I have been developing brain-spine interfaces that control the delivery of epidural electrical stimulation (EES) from brain-decoded commands in adult macaque monkeys. I study the neural basis underlying locomotor activities in motor, premotor, and sensory cortices in monkeys trained to perform different locomotor tasks, and how these can be used to develop decoders that can be used for different, previously untrained tasks. Moreover, I formed part of the team carrying out a Firstin-Man neurorehabilitation program where we used EES of the spinal cord to restore sensation and movement of otherwise paralyzed muscles during standing and walking in people with chronic SCI5. Neural plasticity enabled by EES reinforces a bright future for what was considered, until very recently, an incurable paralysis. However, in order to further accelerate neurological recovery, it is crucial to identigy an individual’s functions that may benefit from activity-based therapy and target neuroprosthetic interventions accordingly. As faculty, my research program will merge the fields of BoMIs and neuroprosthetics to characterize individual function, evaluate candidate interventions and their potential for recovery, and develop personalized neurorehabilitation programs that accelerate and enhance neural plasticity. My research areas will include: 1) predicting recovery and studying its mechanisms after neuromotor disorders, 2) developing control strategies for neuroprosthetics, and 3) continuous adaptation of therapy in individuals with compromised neural systems

Education: MS and PhD | Northwestern University, Biomedical Engineering, 2013 and 2016 BS | The University of Texas at San Antonio, Mechanical Engineering, 2010

Research/Work Experience: Postdoctoral 2016-Present | EPFL, Advisor: Gregoire Courtine (Neuroprosthetics, Brain-Spine Interfaces, Platform for Translational Neurosciences) Graduate 2011-2016 | Northwestern University, Advisor: Ferdinando Mussa-Ivaldi (Motor Learning, Robotics, Rehabilitation) Undergraduate 2009-2010 | UTSA, Advisor: Xiaodu Wang (Hard Tissue Biomechanics) Summer Intern 2009 | Rehabilitation Institute of Chicago, Advisor: Eric Perreault (Neuromuscular Control and Plasticity) Semester Co-op 2008 | Toyota Motor Manufacturing Texas, Plant Engineering (Environmental Engineering)

Selected Publications: 5. Wagner, F.B., … Seáñez, I., et al. “Targeted neurotechnologies restore walking in humans with spinal cord injury.” Nature. 2018. 4. Seáñez, I., et al. “Body-machine interfaces after spinal cord injury: rehabilitation and brain plasticity.” Brain Sciences. 2016. 3. Seáñez, I., et al. “Static vs. dynamic decoding algorithms in a non-invasive body-machine interface.” IEEE Transactions on Neural Systems and Rehabilitation Engineering. 2016. 2. Seáñez, I., et al. “Cursor control by Kalman filter with a non-invasive body-machine interface.” J. Neural Engineering. 2014. 1. Seáñez, I., et al. "A body-machine interface for the control of a 2D cursor." International Conference on Rehabilitation Robotics. 2013.

Awards/Honors: 2018 | Whitaker Concluding Initiative Grant 2016-2018 | Whitaker International Postdoctoral Scholars Award 2014-2016 | NIH Ruth L. Kirschstein National Research Service Awards (F31 – Diversity) 2015 | Northwestern Kellogg School of Management Certificate for Scientists and Engineers 2014 | IEEE EMBS BRAIN Grand Challenges Conference Young Investigator Award - Honorable Mention 2011-2014 | NSF Graduate Research Fellowship Award 2009-2010 | NIH MARC Undergraduate Student Training in Academic Research Award (T34) OR A. SHEMESH, PhD 1MIT, 20 Ames, Cambrighe, MA, 02138, 2MIT Media Lab, MIT, 20 Ames, Cambridge, Massachusetts, 02138 [email protected]

Research Overview: Disease engineering - creating new tools to study brain disease. The cutting edge tools to study the healthy brain can also be applied to study the diseased brain. In particular, physiological tools, such as optogenetic molecules, either stimulate or report activity of neurons of interest. However, as powerful and important as these tools are, in their current form they are mostly suited to study disease after its onset. Therefore most existing tools at the disposal of biomedical investigators are not sufficient to discover the etiology of disease: how does a disease come to pass? While the community that studies the intact brain has seen an abundance of tools that pushed research forward over the past decade, the community that studies brain disease is vastly underserved. To create tools for disease, it would be beneficial to understand the biology of pathology as well as the governing principles of engineering. Since as a PhD student I focused on diseases of the nervous system (working on Alzheimer’s disease), and as a postdoctoral fellow devoted myself to developing tools for research of the brain (working on single resolution readout and control), I will start a first-of-its-kind research field dedicated to creating tools for the study of brain disease: disease engineering. Among these tools will be sensors for the viability states of cells (is a cell healthy, necrotic or apoptotic?), actuators that will switch neurons and brain regions from healthy to a diseased state and back, novel animal models for basic research and drug screening, and devices for interfacing with the nervous system and repairing it. I will combine my experience in protein engineering, nanotechnology, molecular cloning, cell biology and electrical engineering with the unique philosophy for tool building I learned at MIT. This will result in the production of a new set of tools that will enable the community to study disease and develop therapeutics more efficiently. The tools that my group will create will also be relevant to other research fields such as cancer, immunology and infectious or cardiovascular diseases.

Education: 2011 | PhD Neuroscience, 2011, The Hebrew University of Jerusalem. 2010 | MSc Neurobiology, 2009, The Hebrew University of Jerusalem. 2008-2010 | Towards BSc, Electrical Engineering, 2008-2011, Jerusalem College of Engineering. 2004 | BSc Biology (honors program); BSc Psychology, 2004, The Hebrew University of Jerusalem.

Research/Work Experience: 2013-Present | Postdoctoral Fellow; Neroengineering; MIT Media lab and the McGovern Institute for Brain Research; MIT; Advisor: Ed Boyden. 2008-2011 | Graduate Student; Neurobiology; Department of Neurobiology; The Hebrew University of Jerusalem. Advisor: Micha Spira. 2008-2011 | Teaching assistant; Chief TA in Courses in Physiology and Neurobiology. The Hebrew University of Jerusalem. 2004 | Research fellow; Neuro-inflammation and animal behavior; The Hebrew University of Jerusalem. Advisor: Raz Yirmiya.

Selected Publications: • Shemesh OA*., Tanese D*, Zampini V*, Linghu C, Piatkevich KD, Ronzitti E, Papagiakoumou E, Emiliani V† and Boyden ES. †. Temporally precise single-cell-resolution optogenetics. Nature Neuroscience 2017 Dec;20(12):1796-1806. PMID: 29184208. • Shemesh OA.*, Linghu C*, Piatkevich KD, Goodwin D, Freifeld L, Gritton HJ, Romano MF, BenSussen S, Tseng HA, Siciliano CA, Gupta I, Pak N, Young YG, Park W, Keating A, Tye K, Tolias A, Han X, Ahrens M and Boyden ES. †. Precision calcium imaging of dense neural populations via a cell body-targeted calcium indicator. Under Second Revision (Neuron). • Shemesh OA & Spira ME. Rescue of neurons from undergoing hallmark tau-induced Alzheimer's disease cell pathologies by the antimitotic drug paclitaxel. Neurobiology of Disease. 2011 Jul;43(1):163-75. PMID: 21406229. • Shemesh OA & Spira ME. Hallmark cellular pathology of Alzheimer’s disease induced by mutant human-tau expression in cultured Aplysia neurons.. Acta Neuropathologica 2010 Aug;120(2):209-22. PMID: 20422200. • Shemesh OA, Erez H., Gizburg I., Spira ME. Tau-induced microtubules reorientation and displacement leads to axonal traffic jams and neurodegeneration. Traffic, 2008 Apr;9(4):458-71. PMID: 18182010.

Awards/Honors: 2017 - Present | MIT Translational Fellow. 2014-2016 | Simons Foundation Postdoctoral Fellow for Autism Research. 2013 | Edmund and Lily Safra (ELSC) Postdoctoral Fellowship. 2008| Dimitris N. Chorafas Prize 2008, for outstanding work in selected fields in the engineering sciences and medicine. SAGAR SINGH, PhD Bioengineering, University of Pennsylvania, 210 S. 33rd Street, Philadelphia, PA, 19104 . [email protected] Research Overview: My research career has focused on biomechanics and multiscale modeling of traumatic phenomenon, both in the central nervous system, and in the soft tissues of the cervical spine. My pre-doctoral work was performed at Rutgers University, in Dr. David Shreiber’s group in the department of Biomedical Engineering. In my PhD work, I developed novel modeling methodologies in the study of central nervous system injury – particularly in understanding how tissue-level stresses and strains translate to the cellularlevel stresses and strains experienced by axons and glia to determine more accurate mechanical thresholds necessary for primary axonal injury, and to help in designing neural electrodes to mitigate long-term signal attenuation. My decision to join the Spine Pain Research Lab led by Dr, Beth Winkelstein as a postdoctoral researcher was based on the opportunity to apply the methodologies I developed in my PhD work towards a new system; namely the soft tissues of the cervical spine. Under Dr. Winkelstein’s supervision, I have made strides toward becoming an independent researcher, and have acquired a number of new skills in this role including: in vivo studies, survival surgeries, cadveric mechanical testing, and neurobiology. In my time at the Spine Pain Research Lab, I have used an in vitro system to study how the innervated capsular ligament microenvironment can modulate neuronal injury and dysfunction thresholds following supraphysiological loading. Additionally, I helped in developing a model of early-onset osteoarthritis of the facet joints in the cervical spine of rats, and used this system to measure changes to index and adjacent level kinematics to infer the likelihood of traumatic joint injury. Ultimately, I want to further investigate central and peripheral nervous system primary injury pathomechanisms and functional changes that occur in carpal tunnel or brachial plexus trauma, and incorporate the areas of biomechanics and neural engineering at a research and teaching institution looking to develop a more rigorous engineering curriculum.

Education: · Rutgers, The State University of New Jersey, October 2016. PhD, Biomedical Engineering · University of Pennsylvania, May 2008. BSE, Bioengineering

Research/Work Experience: · University of Pennsylvania, Department of Bioengineering, 2016 – present Postdoctoral Researcher, Spine Pain Research Lab; Advisor: Beth A. Winkelstein, PhD · Rutgers, The State University of New Jersey, Department of Biomedical Engineering, 2015 – 2016 Course and Curriculum Designer · Rutgers, The State University of New Jersey, Department of Biomedical Engineering, 2010 – 2016 Graduate Research Assistant; Advisor: David I. Shreiber, PhD · Accenture, 2008 – 2010, Data Archivist Analyst, Systems Integration and Technology · Children’s Hospital of Philadelphia, Department of Pediatric Neurology, 2007 – 2008 Undergraduate Research Assistant; Advisors: Eric Marsh, MD PhD; Brenda Porter, MD PhD

Selected Publications: 1. Singh S, Kartha S, Bulka B, Stiansen N, Winkelstein B. Physiologic facet capsule stretch can induce pain & upregulate MMP-3 in the DRG when preceded by a nonpainful mechanical or chemical exposure. Clinical Biomechanics. 64: 122-130 (2019) 2. Zhang S, Singh S, Winkelstein B. Collagen organization regulates stretch-initiated pain-related neuronal signals in vitro: Implications for structure-function relationships in innervated ligaments. Journal of Orthopaedic Research. 36: 770-777 (2018) 3. Singh S, Pelegri A, Shreiber D. Estimating axonal strain and failure following white matter stretch using contactin-associated protein as a fiduciary marker. Journal of Biomechanics. 14: 1303-1315 (2017) 4. Singh S, Lo M, Damodaran V, Kaplan H, Kohn J, Zahn J, Shreiber D. Modeling the Insertion Mechanics of Flexible Neural Probes Coated with Sacrificial Polymers for Optimizing Probe Design. Sensors. 16: 330-348 (2016) 5. Singh S, Pelegri A, Shreiber D. Characterization of the three-dimensional kinematic behavior of axons in central nervous system white matter. Biomechanics and Modeling in Mechanobiology. 14:1303-1315 (2015)

Awards/Honors: · Rutgers School of Engineering Graduate Fellowship, Rutgers, The State University of New Jersey, 2014 – 2015 · Rutgers TA/GA Professional Development Fund Award, Rutgers, The State University of New Jersey, 2016 – 2016 ALEC SIMON TULLOCH SMITH, PhD Bioengineering, University of Washington, 850 Republican Street, Brotman Building, Room 352, Seattle, WA, 98109 . [email protected]

Research Overview: My research focuses on the development of human stem cell-derived models of peripheral neuropathic disease and the use of these tools to better understand the cellular mechanisms that underpin neuronal and muscle dysfunction in peripheral neuropathy. Specifically, I am focused on generating stem cell-based models of Charcot Marie Tooth disease using CRISPR-Cas9 gene editing and fully characterizing the transcriptome and electrophysiological profile of the resulting cells. In so doing, I aim to better understand how the causal mutation leads to the development of a neurodegenerative phenotype. Additionally, I have been working to compare the in vitro phenotypes of stem cell-derived motor neurons bearing in the TDP-43 protein known to cause amyotrophic lateral sclerosis (ALS) in patients. Through the identification of transcriptomic pathways consistently affected across multiple mutations, I hope to identify gene expression changes that are sufficient for triggering neurodegeneration in ALS. These research areas represent the culmination of my graduate and post-doctoral work and will form the central focus of my independent research laboratory. I also plan to broaden this area of study to encompass the establishment and validation of bioengineered tools for studying neurodegenerative decline in cortical neurons subjected to physical trauma as a model of traumatic brain injury.

Education: -Bachelor of Science in Biology, 2006, Imperial College, London. -Doctor of Philosophy in Tissue Engineering, 2012, University College, London.

Research/Work Experience: -Postdoctoral Research Associate. University of Central Florida, Orlando, FL (2012-2014). -Postdoctoral Fellow. University of Washington, Seattle, WA (2014-2017). -Acting Instructor. University of Washington, Seattle, WA (2017-present).

Selected Publications: Full bibliography available at Google Scholar (https://scholar.google.com/citations?user=WuJbC38AAAAJ&hl=en) or PubMed (https://www.ncbi.nlm.nih.gov/myncbi/1-CLeoPclyyQl/bibliography/public/) -Natarajan A., Smith A.S.T. (co-first author), Berry B.J., Lambert S., Molnar P., Hickman J.J. (2018) Temporal Characterization of Neuronal Migration Behavior on Chemically Patterned Neuronal Circuits in a Defined In Vitro Environment. ACS Biomaterials Science and Engineering. 4(10): 3460-3470. -Smith A.S.T., Yoo H., Yi H., Ahn A.H., Lee J., Shao G., Nagornyak E., Laflamme M.A., Murry C.E., Kim D.H. (2017) Micro- and Nano-Patterned Conductive Graphene-PEG Hybrid Scaffolds for Cardiac Tissue Engineering. Chem Comms. 53: 7412-7415. PMID: 28634611. -Smith A.S.T., Passey S.L., Martin N.R., Player D.J., Mudera V., Greensmith L., Lewis M.P. (2016) Creating Interactions Between Tissue-Engineered Skeletal Muscle and the Peripheral Nervous System. Cells Tissues Organs. 202(3-4):143-158. PMID: 27825148. -Smith A.S.T., Long C.J., Pirozzi K., Hickman J.J. (2013) A Functional System for High-Content Screening of Neuromuscular Junctions In Vitro. Technology (Singap World Sci). 1(1):37-48. PMID: 25019094. -Smith A.S.T., Passey S., Greensmith L., Mudera V., Lewis M.P. (2012) Characterization and Optimization of a Simple, Repeatable System for the Long-Term In Vitro Culture of Aligned Myotubes in 3D. J Cell Biochem. 113(3):1044-53. PMID: 22065378.

Awards/Honors: -Medical Research Council Doctoral Training Award - Full Scholarship for Graduate School (2007-2011). -University of Central Florida Commercialization Grant 1055766 (Role: Co-Investigator, 2013). -NIH T32 Training Grant Awardee (2015-2016). -UW Bioengineering Annual Retreat. Best Poster Award (2016). -UW Innovator of the Year. Innovator Showcase Award (2016). -NIH SBIR Phase 1 Award. R43 HL131169-01 (Role: PI, 2016-2017) -Jaconette L. Tietze Young Scientist Award (2017). -UW Distinguished Teaching Award Nominee (2017). -UW Institute for Translational Health Science KL2 Career Development Award. NIH KL2 TR002317 (Role: PI, 2017-2020) -NIH UG3 EB028094 - Chips In Space Program (Role: Co-Investigator, 2018-2020). -Axion Biosystems Travel Award to Attend BMES Annual Conference (2019). QUINTON SMITH, PhD The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142 [email protected]

Research Overview: As the field of tissue engineering evolves, human induced pluripotent stem cells (hiPSCs) have begun to address the limitations of cell sourcing and the fabrication of synthetic materials has introduced bio-compatible scaffolds, that can be engineered to meet the mechanical and biochemical complexity of native tissue. The marriage of these technologies permits the delivery of immunologically competent cells with designer extracellular matrices (ECM), that can act in concert to augment and replace diseased tissue in vivo. However, the clinical success of tissue-engineered constructs hinges upon a functional vascular bed, which acts to not only supply oxygen and nutrients, but also to remove waste. My graduate research implemented engineering tools, namely high-throughput image analysis, tunable biomaterials, micropatterning and microfluidics, to investigate the role of various biophysical stimuli on hiPSC gastruloid self- organization, mesoderm specification, and downstream vascular differentiation and maturation. My postdoctoral work focuses on leveraging these tools to construct multi-cellular architectures for facilitating liver regeneration. As a principal investigator, my lab will utilize a highly interdisciplinary approach to direct multi-scale integration of hiPSC-based vasculature with tissue-specific organoids, focused on the controlled delivery of mechanical cues.

Education: Ph.D. in Chemical and Biomolecular Engineering, Johns Hopkins University (2017) B.S. in Chemical Engineering, University of New Mexico (2011)

Research/Work Experience: Postdoctoral Fellow, Massachusetts Institute of Technology, Koch Institute for Integrative Cancer Research, Advisor: Dr. Sangeeta Bhatia (2017- Present) Graduate Research Assistant, Johns Hopkins University, Department of Chemical and Biomolecular Engineering, Advisor: Dr. Sharon Gerecht (2012-2017) Technician, Sandia National Laboratories, Biodefense and Emerging Infection Diseases, Advisor: Dr. Jerilyn Timlin (2011-2012) Student Technician, University of New Mexico, Epidemiology, Advisor: Dr. Marianne Berwick (2007-2011)

Selected Publications: Smith Q., Macklin B., Chan XY., Jones H., Trempel M., Gerecht, S. “Differential HDAC6 activity modulates ciliogenesis and subsequent mechanosensing of endothelial cells derived from pluripotent stem cells.” Cell Reports (2018). Smith, Q.*, Rochman N.*, Carmo AM., Vig D., Chan, X. Y. Sun S., Gerecht, S. “Cytoskeletal tension regulates mesodermal spatial organization and subsequent vascular fate.” PNAS (2018). *co-authorship Smith, Q. & Gerecht, S. “Extracellular Matrix Regulation of Stem Cell Fate.” Curr Stem Cell Rep (2018) 4: 13. https://doi.org/10.1007/s40778-018-0111-2 Smith, Q., Chan, X. Y., Carmo, A. M., Trempel, M., Saunders, M., & Gerecht, S. (2017). Compliant substratum guides endothelial commitment from human pluripotent stem cells. Science Advances, 3(5), e1602883. http://doi.org/10.1126/sciadv.1602883 Smith, Q., Stukalin E., Kusuma S., Gerecht, S., Sun, S. “Stochasticity and Spatial Interaction Govern Stem Cell Differentiation Dynamics.” Scientific Reports (2015) 5; 12617-10. Smith Q., Gerecht S. “Going with the flow: microfluidic platforms in vascular tissue engineering.” Current Opinion in Chemical Engineering (2014) 42–50.

Awards/Honors: Howard Hughes Medical Institute Hanna Gray Fellow, $250,000/year up to four years as PI (2018 – Current) US Patent 0216063, Three-dimensional vascular network assembly from induced pluripotent stem cells (2018) Siebel Scholar, Awarded Annually for Academic Excellence and Demonstrated Leadership to Over 90 Top Students from World’s Leading Graduate Schools (2017) F31 NIH/NHLBI Ruth L. Kirschstein NRSA Predoctoral Fellowship (2016 – 2017) NSF Graduate Research Fellowship Program (2013 – 2016) BMES Innovation and Career Development Award (2014) GRC Poster Award: Signal Transduction in Engineered Extracellular Matrices (2014) M. HONGCHUL SOHN, PhD Physical Therapy and Human Movement Sciences, Northwestern University, 645 North Michigan Avenue, Chicago, IL, 60611 . [email protected]

Research Overview: My primary research goal is to understand how the human nervous system controls and learns purposeful motor actions, and how that ability is altered by neuromuscular injury and disease. In long-term, this work is aimed at developing enhanced neurorehabilitation strategies for individuals with compromised sensorimotor function after neurological injury. As a mechanical engineer uniquely trained at the interface of neuroscience and rehabilitation, my work integrates engineering principles of mechanics and system dynamics & control to characterize the neurophysiological mechanisms underlying human movement. My doctoral work focused on theoretical investigation of neural control for posture and movement using musculoskeletal modeling and simulation. Importantly, predictions made possible by novel computational tools I developed could aid the interpretation of individual differences in motor control strategy and compensation following neuromuscular deficits. In my early postdoctoral training, I have expanded my expertise in experimental methods for using neurophysiological measurements, virtual reality, and robotic systems to probe into sensory feedback mechanisms that are vital to motor learning. Recently, I have started investigating structural alterations in muscles post-stroke using imaging techniques. This work seeks to unravel the complicated interaction between neural deficits and secondary musculoskeletal changes, which confounds the treatment of functional impairment in chronic stroke survivors. Leveraging these experiences and skills, future research in my lab will combine computational and experimental approaches to: 1) elucidate sensorimotor mechanisms that facilitate learning/recovery of functional movement and 2) identify motor control strategies to mitigate/prevent musculoskeletal complications after neural injury. My research will create the foundational knowledge necessary to develop novel therapeutic interventions, initially targeted at survivors of stroke and spinal cord injury. These results will also have broad impact for applications beyond rehabilitation, including the design of human-machine interfaces and training programs for medical, military, and athletic purposes.

Education: 2009 – 2015 PhD, Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 2002 – 2006 BS, Mechanical and , Seoul National University, Seoul, Korea

Research/Work Experience: 2018 – present Postdoctoral Scholar, Physical Therapy and Human Movement Sciences, Northwestern University, Advisors: Julius Dewald and Wendy Murray 2015 – 2018 Postdoctoral Fellow, Biomedical Engineering, Northwestern University, Advisors: Eric Perreault and James Patton 2010 – 2015 Graduate Research Assistant, Mechanical Engineering, Georgia Institute of Technology, Advisor: Lena Ting 2010 – 2010 Graduate Teaching Assistant, Mechanical Engineering, Georgia Institute of Technology, Instructor: Steven Johnson 2006 – 2009 Research Officer, Department of Propulsions, Aero Technology Research Institute of Republic of Korea Air Force

Selected Publications: • Sohn MH, Smith DM, Ting LH (2019). Effects of kinematic complexity and number of muscles on musculoskeletal model robustness to muscle dysfunction. PLOS ONE 14(7): e0219779. • Sohn MH, Baillargeon EM, Lipps DB, Perreault EJ (2017). Stretch reflexes in shoulder muscles are described best by heteronymous pathways. Converging Clinical and Engineering Research on Neurorehabilitation II. Biosystems & Biorobotics, vol 15. Springer. • Sohn MH, Ting LH (2016). Suboptimal muscle synergy activation patterns generalize their motor function across postures. Frontiers in Computational Neuroscience doi:10.3389/fncom.2016.00007. • Sohn MH, McKay JL, Ting LH (2013). Defining feasible bounds on muscle activation in a redundant biomechanical task: practical implication of redundancy. Journal of Biomechanics 46(7): 1363-1368. • Sohn MH, Ting LH (in revision, submitted to PLOS Computational Biology). The cost of being stable: Trade-offs between effort and stability across the landscape of redundant motor solutions.

Awards/Honors: 2019 Invited Travel Award to OpenSim Advanced User Workshop, National Center for Simulations in Rehabilitation Research 2018 Scholarship Award, Society for the Neural Control of Movement 2017 NIH NRSA Individual Postdoctoral Fellowship (F32) 2015 NIH NRSA Institutional Training Grant Appointee (T32), Northwestern University 2015 Finalist in Poster Competition, Summer School on Neurorehabilitation KIMBERLY A. STEVENS, PhD Biomedical Engineering, Purdue, 40 Cavalry Ct, West Lafayette, IN, 47906 [email protected]

Research Overview: Blood transport in the human body is one of the most intimate, relevant, and complex fluids problems in existence, given that continuous functioning of complex, non-linear, pulsatile flows of a non-Newtonian fluid throughout tens of thousands of miles of flexible, variable sized conduit universally separates humans from life and death. In my research at Purdue University as a Lillian Gilbreth Postdoctoral Fellow, I seek to fuse commonly used medical imaging data of multiple modalities (e.g. digital subtraction angiography, 4D flow MRI, dynamic susceptibility contrast MR perfusion, etc.) with image-based computational fluid dynamics models to create higher-fidelity cerebrovascular flow models. The models can be used to improve fundamental understanding of cardiovascular disease pathogenesis, develop improved cardiovascular risk assessment models, and inform patient-specific clinical decisions; applying the enhanced models in these ways will be the initial focus of my research program as a faculty member. During Postdoctoral Fellowship, I developed collaborations with clinicians I will continue to leverage as a faculty member. My doctoral research involved experimental and numerical phase-change heat transfer modeling. During my Masters, I created in vitro self oscillating vocal fold models. My background in thermal-fluid transport, computer vision, and data analysis together with the expertise and collaborations I developed during my post-doc have given me a unique set of tools to develop solutions to cardiovascular flow problems at the interface of mechanical engineering, biomedical engineering, and medicine.

Education: • PhD, Mechanical Engineering, 2018, Brigham Young University • MS, Mechanical Engineering, 2015, Brigham Young University • BS, Mechanical Engineering, 2012, Brigham Young University

Research/Work Experience: • Lillian Gilbreth Postdoctoral Research Fellow, Purdue University. Advisors: Drs. Ivan Christov and Vitaliy Rayz. 2018-2020. • Doctoral Research Assistant, Brigham Young University. Advisor: Dr. Brian Iverson. 2014-2018 • Summer Scholar, Air Force Research Lab. Advisor: Brent Taft. 2016 • National Science Foundation Fellow, Ritsumeikan University, East Asia Pacific Summer Institute Fellowship. Advisor: Dr. Isao Tokuda. 2014 • Research Assistant, Brigham Young University. Advisor: Dr. Scott Thomson. 2011-2014. • Undergraduate Research Assistant, Brigham Young University. Advisor: Dr. David Fullwood. 2007-2009.

Selected Publications: 1. Stevens, KA, Smith, SM, Taft, BS. 2019. Variation in Oscillating Heat Pipe Performance. Applied . 149:987-995. 2. Stevens, KA, Esplin, CD, Davis, TM, Butterfield, DJ, Ng, PS, Bowden, AE, Jensen, BD, Iverson, BD. 2018. Superhydrophobic, Carbon-Infiltrated Carbon Nanotubes on Si and 316L Stainless Steel with Tunable Geometry. Applied Physics Letters. 112:211602. 3. Stevens, KA, Crockett, J, Maynes, RD, and Iverson, BD. 2017. Two-phase flow pressure drop in superhydrophobic channels. International Journal of Heat and Mass Transfer. 110:515-522. 4. Stevens KA, Jette ME, Thibeault SL, Thomson SL. 2016. Quantification of Porcine Vocal Fold Geometry. Journal of Voice. 30(4):416-426. 5. Stevens KA, Shimamura R, Imagawa H, Sakakibara K, Tokuda IT. 2016. Validating Stereo-Endoscopy with a Synthetic Vocal Fold Model. Acta Acustica united with Acustica. 102(4):745-751(7).

Awards/Honors: • Lillian Gilbreth Postdoctoral Fellowship, Purdue University, 2018-2020 • Best Poster - Advancing Health: An Engineering Medicine Partnership Symposium, 2019 • Advanced Studies Institute, Los Alamos National Lab, 2018 • 2nd Place, BYU College of Engineering 3 Minute Thesis Competition, 2017 • Air Force Research Lab Outstanding Scholar Award, 2016 • East Asia Pacific Summer Institute Fellowship, NSF, 2014 SHAWANA TABASSUM, PhD Electrical and Computer Engineering, Iowa State University, 3131 Coover Hall, Iowa State University, Ames, IA, 50014 [email protected] Research Overview: My research is mainly focused on developing in-vivo and point-of-care optical sensors and systems at nanoscale for biomedical applications. My sensors have applications also in sustainable agriculture and field/farm ecosystem monitoring and management. First, I developed an integrated dual-modality microfluidic sensor for a cancer biomarker detection using lithographic plasmonic crystal. I demonstrated a femtomolar level limit of detection for quantifying a breast cancer biomarker, which is clinically important for early-stage cancer diagnosis. The promising outcome of this work, as reflected in the submission of a patent application (ISURF 4729), encouraged me to discover further potentials of my devices. Second, considering the recently discovered immense therapeutic potential of hydrogen gas, I developed a plasmonic crystal-based high- sensitivity hydrogen gas sensor. The results form the basis for realizing an in-vivo optical resonant device for sensitive monitoring of hydrogen in organs upon administration, to unravel the mechanisms underlying its therapeutic potential. I have an accepted poster in 2019 BMES Annual Meeting on this work with paper ID #2721. Apart from these biosensing research, I have continuously sought inputs from the experts in biological/medical domain and I have successfully established 3 significant research collaborations: a) with a world renowned physician in the Department of Medicine, University of Iowa Medical School, in developing a label-free, minimally invasive glucose monitoring sensor and a closedloop feedback model to achieve homeostatic metabolic control of glucose (third); b) with a professor at Rutgers University and his clinician colleagues in developing a point-of-care biochip for improved and rapid diagnosis of sepsis (fourth); c) We are also evolving collaborations with a faculty from genetics, development and cell biology to look for collaborations on providing nanostructured platform for stem cell growth and to use fiber-optics to investigate nervous system repair (fifth). I have also completed ISU’s Innovation Corps Site program as the Entrepreneurial Lead to gain training on technology transfer of my devices. I also developed a guided mode resonance nanostructure on the tip of an optical fiber, coated with Cu complex and graphene oxide to respectively detect gaseous ethylene (plant stress indicator) and NH3. A patent application has been submitted for these sensors (ISURF 4453). We are currently working on advancing the technology to its next step of fabricating a fully-integrated, wireless, multiplexed gas- sensing system, ready for field use, to assist the farmers in agro- and farm-ecology management practices including nitrogen, pesticide, and other farm management.

Education: Ph.D. in Electrical Engineering, December 2018, Iowa State University (ISU), Ames, IA, USA, B.Sc. in Electrical Engineering, May 2014, Bangladesh University of Engineering and Technology (BUET), Dhaka, Bangladesh

Research/Work Experience: Postdoctoral Research Fellow – Health & Environmental Monitoring-- December 2018 – Present Research Intern, NSF-INTERN Supplement, Ames, IA--November 2018 – Present Graduate Research Assistant – Microelectronics and Photonics--August 2014 – December 2018

Selected Publications: 1. Ali, M. A. Tabassum, S. et al. Integrated dual-modality microfluidic sensor for biomarker detection using lithographic plasmonic crystal, Lab Chip 2018 2. Tabassum, S. et al. Determination of dynamic variations in optical properties of graphene oxide in response to gas exposure based on thin-film interference, Opt. Express 2018. 3. Tabassum, S. et al. Nanopatterned optical fiber tip for guided mode resonance and application to gas sensing, IEEE Sens. J. 2017. 4. Tabassum, S. et al. Plasmonic crystal-based gas sensor toward an optical nose design, IEEE Sens. J. 2017.

Awards/Honors: • Career Development Award, by Biomedical Engineering Society (BMES)--July 2019 • Awarded $3k by ISU’s Innovation-Corps Site program for furthering entrepreneurship--March 2019 • Awarded NSF-INTERN Supplement ($50k)--November 2018 • ISU Research Excellence Award ($500)–awarded to the top 10% of graduate students for excellence in research--June 2018 • Invitation to highlight my work on the cover of the Lab-on-a-chip journal--February 2018 • Excellent Reviewer recognition from the journal 'Sensor Letters'--January 2018 • Best Paper Award Finalist in IEEE Sensors Conference 2016--September 2016 • Dean’s List Award and Merit Scholarship, BUET–awarded to the top 10% students for excellent academic performance-- 2009-2014 GULISTAN TANSIK, PhD Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL, 33146 [email protected]

Research Overview: My academic training and research experience have provided me with an excellent background in multiple scientific disciplines, including molecular biology, chemistry, materials science, and nanotechnology. My PhD research focused on the design of biomimetic hydrogel scaffolds for in vivo biomineralization and the regeneration of bone tissue. I demonstrated that peptide nanofibers that mimic glycosaminoglycans are promising scaffolds for osteogenic differentiation of rat mesenchymal stem cells and in vivo bone regeneration. In addition, I established that the stiffness changes in stem cells throughout osteogenic differentiation are strongly affected by the degree of biomineralization. During my master’s studies, I fabricated noncytotoxic PLGA coated iron oxide nanoparticles (diameter < 100nm) with superparamagnetic characteristics that could be loaded with an anti-cancer drug. I demonstrated the in vitro efficacy of the drug laden nanoparticles. Currently, as a postdoctoral researcher in the Physiomimetic Microsystems Laboratory, I am developing in vitro microphysiological systems to model in vivo microenvironments by incorporating biochemical and mechanical cues. I have developed a dynamically stiffening hydrogel platform to promote maturation of stem cellderived cardiomyocytes. Simultaneously, I have built an in vitro cardiac amyloidosis disease model on a chip. For my future career, I want to leverage my expertise in developing new biomaterial based physiomimetic platforms, with the goal of understanding the underlying mechanisms of bone and cardiac diseases and developing effective treatments for them.

Education: PhD Materials Science and Nanotechnology 2017, Ihsan Dogramaci Bilkent University, Ankara-Turkey. MSc Biology Middle East Technical University 2012, Ankara-Turkey. BSc Biology Middle East Technical University 2008, Ankara-Turkey.

Research/Work Experience: 2017-present: Postdoctoral Research Associate, Department of Biomedical Engineering, University of Miami, FL. Advisor: Dr. Ashutosh Agarwal Fall 2018: Guest Lecturer, ECE543/643 BioNanotechnology and BME 565 Cell & Tissue Engineering, University of Miami, FL Spring 2019: Guest Lecturer, BME 335 Biomaterials, University of Miami, FL 2012-2017: Graduate Research Assistant, Department of Materials Science and Nanotechnology, Ihsan Dogramaci Bilkent University, Ankara-Turkey. Advisors: Dr. Mustafa Ozgur Guler & Dr. Ayse Begum Tekinay. 2008-2012: Graduate Research Assistant, Department of Biology, Middle East Technical University, Ankara-Turkey. Advisor: Dr. Ufuk Gunduz. August 2007 - October 2007: Research Intern, The Scientific and Technological Research Council of Turkey, Marmara Research Center, Genetic Engineering and Biotechnology Institute, Gebze, Turkey

Selected Publications: 1) Tansik G, Kilic E, Beter M, Demiralp B, Sendur GK, Can N, Ozkan H, Ergul E, Guler MO, Tekinay AB. A glycosaminoglycan mimetic peptide nanofiber gel as an osteoinductive scaffold. Biomaterials Science, 2016, 4, 1328 - 1339, doi:10.1039/C6BM00179C. 2) Topal AE*, Tansik G*, Ozkan AD, Guler MO, Tekinay AB, Dana A. Nanomechanical characterization of osteogenic differentiation of mesenchymal stem cells on bioactive peptide nanofiber hydrogels. 2017, Advanced Materials Interfaces, Wiley, DOI:10.1002/admi.201700090. (*equal contribution) 3) Eren ED*, Tansik G*, Tekinay AB, Guler MO. Mineralized peptide nanofiber gels for enhanced osteogenic differentiation. ChemNanoMat, 2018, doi:10.1002/cnma.201700354. (*equal contribution) 4) Tansik G, Yakar A, Gunduz U. Tailoring magnetic PLGA nanoparticles suitable for doxorubicin delivery. Journal of Nanoparticle Research. 2013, 16:2171. 5) Tansik G, Alassaf A, Velluto D, Prabhakar R, Agarwal A. Modeling cardiac amyloidosis disease on a chip. (in preparation)

Awards/Honors: 2010-2012: The Scientific and Technological Research Council of Turkey (TUBITAK), Scientific and Technological Research Projects (1001) Program Graduate Scholarship, Middle East Technical University, Ankara, Turkey 2014-2017: TUBITAK-2211-Graduate Scholarship Program for Primary Fields Scholarship, Ihsan Dogramaci Bilkent University, Ankara, Turkey BRITTANY TAYLOR, PhD Orthopaedic Surgery, University of Pennsylvania, 3450 Hamilton Walk, 371 Stemmler Hall, Philadelphia, PA, 19104 [email protected]

Research Overview: My overall research focus is on tailored musculoskeletal tissue engineering approaches to synergistically complement and improve the native healing environment. As a postdoctoral researcher in the McKay Orthopaedic Research Laboratory at the University of Pennsylvania, my research objective is to elucidate the biological and mechanical implications of localized non- steroidal antiinflammatory drug (NSAID) delivery in a rat supraspinatus tendon (rotator cuff) injury and repair model. This project aims to address the high prevalence of tendon re-tear following repair by implanting a controlled bilayer delivery system (BiLDS) to promote improved tendon healing by modulating inflammation. The BiLDS remained intact up to 8 weeks at the repair site, mitigated the inflammatory response over time, and led to the recovery of the tendon’s structural properties. My graduate research was on the development of an osteoinductive pre-vascularized composite scaffold for significant bone loss. I developed an innovative scaffold that mimicked both the porous trabecular bone and highly-dense coritcal bone structures, imultaneously promoted osteogenic and angiogenic differentiation of human mesenchymal stem cells, and significantly increased subcutaneous cellular infiltration and neovascularization without the addition of growth factors. This collection of work exploits the ability of these transformative technologies to provide physical and chemical cues for improved musculoskeletal tissue repair. My future research interest is investigating cell-free strategies, such as extracellular vesicles (EVs), for enhanced tissue healing. EVs are cellular-derived vesicular structures that play a critical role in many diseases and tissue regeneration following injury. My independent project (recently funded by the Burroughs Wellcome Fund) will define the therapeutic potential of EVs by evaluating their healing response on tendon repair and maintenance in a rat supraspinatus tendon injury and repair model. This work will provide significant contributions to the musculoskeletal tissue regeneration field and serve as the foundation for broader research questions, such as: (1) what is the mechanistic role of EVs in musculoskeletal tissue healing, and (2) can EVs be utilized as drug-carriers for improved tissue repair?

Education: • Ph.D., Biomedical Engineering, 2016, Rutgers University, New Brunswick, NJ (Thesis Advisor: Dr. Joseph Freeman) • B.S., Biomedical Engineering, 2010, University of Virginia, Charlottesville, VA

Research/Work Experience: • University of Pennsylvania Provost's Postdoctoral Fellow, Aug. 2016 – Current (Advisor: Dr. Louis Soslowsky) • Graduate Research Fellow, Rutgers University (New Brunswick), Aug. 2011 – April 2016 • Graduate Research Intern, Research and Discovery Team at Celgene Cellular Therapeutics, May - Aug. 2015 • Post-Baccalaureate Research Assistant, Virginia Polytechnic Institute and State University (Virginia Tech), Aug. 2010 – June 2011 • Undergraduate Research Assistant, University of Virginia, June 2008 - May 2010 (Advisor: Dr. Edward Botchwey)

Selected Publications: 1. Taylor, B.L., Indano, S., Yankannah, Y., Patel, P., Perez, I., Freeman, J.W. Decellularized Cortical Bone Scaffold Promotes Organized Neovascularization In Vivo. Tissue Engineering, Part A. Tissue Engineering, Part A. 25(13-14):p. 964-977 (2019) 2. Taylor, B.L., Cheema, A., Soslowsky, L. Tendon Pathology in Hypercholesterolemia and Familial Hypercholesterolemia. Current Rheumatology Reports. 19 (76): p. 1-6 (2017) 3. Taylor, B.L., Limaye, A.N., Yarborough, J.A., Freeman, J.W. Investigating Various Processing Techniques for a Bovine Gelatin Scaffold for Bone Tissue Regeneration. Journal for Biomedical Research: Part B. 105(5): p. 1131-1140 (2016) 4. Taylor, B. *, Andric, T. *, Degen, K., Whittington, A., Freeman, J. Fabrication and Characterization of Three Dimensional Electrospun Cortical Bone Scaffolds. Nanomat. Environ. 2: p. 13-21 (2014) (*Equal contribution) 5. Taylor, B.L., Andric, T., Freeman, J.W., Recent Advances in Bone Graft Technologies. Recent Patents on Biomedical Engineering. 1:p. 1-7 (2013)

Awards/Honors: • Burroughs Wellcome Fund Postdoctoral Enrichment Program Award, 2019 - 2022 • Diversity Supplement Postdoctoral Fellowship (NIH/NIAMS), 2019 - 2022 • 2018 Mid-Atlantic PREP/IMSD Research Symposium (MAPRS) Distinguished Alumni Award, 2018 • NIH T32 Biotechnology Training Predoctoral Fellowship (T32 GM8339), 2014 - 2016 • NSF Alliances for Graduate Education and the Professoriate (AGEP) Fellowship, 2014 - 2016 • National Science Foundation Graduate STEM Fellowship in K-12 Education (GK-12) 2013 - 2014 KAYVAN FOROUHESH TEHRANI, PhD Regenerative Bioscience Center, University of Georgia, 425 River rd., Athens, Georgia, 30602 - [email protected]

Research Overview: The overarching goal of my research is to develop methods for super-resolution deep tissue intravital microscopy, with the end goal of studying neural regulation of the skeletal system. I received my doctoral training at the laboratory of Dr. Peter Kner, one of the pioneers of structured illumination microscopy. There, I developed methods for deep-tissue super- resolution microscopy inside of the brain of Drosophila melanogaster, using the Adaptive Optics (AO) technology [1]. I further researched on employing quantum dots (QD) for STORM super-resolution microscopy; exploiting the inherent high brightness of QDs to improve the resolution of the imaging technique [2]. After concluding my doctoral research, I joined Dr. Luke Mortensen’s lab as a postdoctoral associate, where I am currently gaining experience in intravital microscopy. My current research interests include AO technology for deep tissuemicroscopy inside of bone marrow cavity. I have developed methods to model wavefront error when samples inside of the bone are imaged [3]. I have further extended our microscopy capability to perform fast volumetric imaging [4]. I am also interested in studying musculoskeletal diseases, and the role mitochondria play in bone and muscle regeneration [5, 6]. As an independent researcher, my laboratory would primarily focus on: 1) developing innovative methods for AO mediated deep tissue microscopy, 2) two and three-photon super-resolution intravital microscopy, 3) study the neural regulation of bone homeostasis and regeneration for therapeutic purposes.

Education: 2015 - Ph.D. - Biological Engineering - University of Georgia, United States 2009 - M.Sc. - Optical Engineering - University of Nottingham, Nottingham, United Kingdom 2006 - B.Eng. - Eletronics Engineering - Azad University, Tehran, Iran

Research/Work Experience: 2015 – present Post-doctoral research associate, University of Georgia, Athens, GA 2012 – 2015 Pre-doctoral research assistant, University of Georgia, Athens, GA 2006 – 2008 Research and Development Engineer, Baset Pazhoh Novin Ltd., Tehran, Iran 2003 – 2006 Research and Development Technician, Professor Hessaby Foundation, Tehran, Iran

Selected Publications: [1]K. F. Tehrani, J. Xu, Y. Zhang, P. Shen, and P. Kner, "Adaptive optics stochastic optical reconstruction microscopy (AO-STORM) using a genetic algorithm," Optics Express, vol. 23, pp. 13677-13692, 2015. [2]J. Xu, K. F. Tehrani, and P. Kner, "Multicolor 3D Super-resolution Imaging by Quantum Dot Stochastic Optical Reconstruction Microscopy," ACS Nano, vol. 9, pp. 2917-2925, 2015. [3]K. F. Tehrani, P. Kner, and L. J. Mortensen, "Characterization of wavefront errors in mouse cranial bone using second- harmonic generation," Journal of Biomedical Optics, vol. 22, pp. 036012-036012, 2017. [4]K. F. Tehrani, C. V. Latchoumane, W. M. Southern, E. G. Pendleton, A. Maslesa, L. Karumbaiah, et al., "Five- dimensional twophoton volumetric microscopy of in-vivo dynamic activities using liquid lens remote focusing," Biomedical Optics Express, vol. 10, pp. 3591-3604, 2019. [5]K. F. Tehrani, E. G. Pendleton, W. M. Southern, J. A. Call, and L. J. Mortensen, "Two-photon deep-tissue spatially resolved mitochondrial imaging using membrane potential fluorescence fluctuations," Biomedical Optics Express, vol. 9, pp. 254- 259, 2018. [6]W. M. Southern, A. S. Nichenko, K. F. Tehrani, M. J. McGranahan, L. Krishnan, A. E. Qualls, et al., "PGC-1α overexpressionpartially rescues impaired oxidative and contractile pathophysiology following volumetric muscle loss injury," Scientific Reports, vol.9, p. 4079, 2019.

Awards/Honors: NIH K99/R00 application under review B.P. Verma award for scientific and leadership excellence, University of Georgia College of Engineering (2015) Excellence in mentorship, NSF REU program (2018) Recognition for great contribution to career development of the University of Georgia graduates (2019) SPIE travel award (2015) Best poster award, College of Engineering, University of Georgia (2014)

MARTIN TRAPECAR, PhD Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139 . [email protected]

Research overview As a trained immunologist and bioengineer in the lab of Linda G. Griffith at MIT, I am developing human physiomimetic technologies of organ-organ interactions to decipher fundamental drivers of tissue homeostasis and immunometabolic diseases. In collaboration with the team of prof. Rudolf Jaenisch at the Whitehead Institute, I have created a human physiomimetic model of the gut -liver-brain axis that includes components of both the innate and adaptive immune system. Using this technology, I investigated the importance of organorgan interactions for systemic homeostasis and further applied this approach to study the entanglement between microbiome-derived short-chain fatty acids (SCFA) and CD4+ Treg/Th17 cells in ulcerative colitis (in review) as well as Parkinson’s disease (in review). By employing a multiomics and systems immunology approach, in collaboration with Prof. Douglas Lauffenburger and his team, I hav e uncovered the ability of SCFA to exacerbate both autoimmunity and neuronal pathology via metabolic reprograming in T cell-dependent as well as independent ways. These proof-of-principle studies underpin the emerging utility of physiomimetic modeling in combination with systems biology to investigate mechanistic causality in multifactorial diseases and in the same time to learn about fundamental drivers of tissue homeostasis. As an independent investigator, I aim to build upon these findings and to further develop this novel system in combination with systems biology to establish interaction networks that will allow me to identify mechanistic causality behind gutliver- brain immunometabolic pathologies, to identify tangible targets for disease amelioration and identify laws governing organ-organ quilibrium with implications for regenerative medicine.

Research/Work Experience • Department of Biological Engineering, Massachusetts Institute of Technology, January 2018-Present Postdoctoral Associate, Advisor: Prof. Linda G. Griffith, PhD • Gladstone Institutes of Virology and Immunology/ UCSF, September 2014- December 2017 Postdoctoral Fellow, Advisor: Prof. Shomyseh Sanjabi, PhD • Biolog d.o.o., 2011- 2014; Co-founder, CEO • Medical Faculty, University of Maribor, 2010-2014 Graduate researcher/Teaching Assistant, Advisors: Prof. Avrelija Cencic, PhD; Prof. Marjan Slak Rupnik, PhD

Education • University of Maribor, Medical Faculty, PhD, Biomedical Technology, September 2014 • University of Maribor, Medical Faculty, MSc, Food Safety, September 2010 • University of Maribor, Faculty of Agriculture and Life Sciences, Uni. Dipl. Ing. (MSc equivalent), September 2009

Selected Publications • Trapecar, M, Communal, C, P, Velazquez, J, Maass, CA, Huang, Y, Schneider, K, Wright, CW, Eng, G, Yilmaz, O, Trumper, D, Griffith, LG. Gut-Liver physiomimetics reveal paradoxical modulation of IBD-related inflammation by short-chain fatty acids. (2019) – In review, Deposited at BioRxiv • Trapecar, M, Khan, S, Cohn, BL, Wu, F, Fontaine, KA, Ott, M, Sanjabi, S. B cells are the predominant mediators of early systemic viral dissemination during rectal LCMV infection. Mucosal Immunology doi:10.1038/s41385-018-0009-4 (2018) • Trapecar, M, Khan, S, Roan, N, Chen, TH, Telwatte, S, Deswal, M, Pao, M, Somsouk, M, Deeks, SG, Hunt, PW, Yukl, S, Sanjabi, S. An Optimized and Validated Method for Isolation and Characterization of Lymphocytes from HIV+ Human Gut Biopsies. AIDS Research and Human Retroviruses doi.org/10.1089/AID.2017.0208. (2017) • Trapecar*, M, Goropevsek, A, Gorenjak, M, Gradisnik, L, Rupnik, M. A co-culture model of the developing small intestine offers new insight in the early immunomodulation of enterocytes and macrophages by lactobacillus spp. through Stat1 and Nf-kB p65 translocation. Plos One, 9(1), 1-8. (2014) *Corresponding Author • Sarenac, T, Trapecar*, M, Gradisnik, L, Slak Rupnik, M, Pahor, D. Single-cell analysis reveals IGF-1 potentiation of inhibition of the TGF-β/Smad pathway of fibrosis in human keratocytes in vitro. Scientific Reports 6: 34373. (2016) *Corresponding Author

Awards Merck Exploratory Science Fellowship 2019-2020; SMI Young Investigator Award 2017; Gladstone career Advancement Award 2017; Slovenian Young Researcher PhD Scholarship 2010-2014; Vet&Agro Undergraduate stipend 2004-2006 VALERIE TUTWILER, PhD Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, Room 1149, Philadelphia, PA, 19104 . [email protected]

Research Overview: Fibrin is an extracellular matrix protein and a major component of blood clots that stem bleeding (hemostasis) and thrombi that block vessels leading to deleterious conditions such as heart attacks and strokes (thrombosis). Moreover, the unique mechanical, structural, and biocompatibility properties of fibrin make it a versatile biomaterial. Understanding the role of fibrin in the regulation of bleeding and thrombosis is a complex problem but has the potential to influence the development of biomaterials and targeted therapeutics for the treatment of bleeding and thrombotic conditions. Fibrin lies at the intersection of coagulation and inflammation; however, the key mechanisms governed this important relationship remain elusive. The expected directions of my future laboratory are 2) develop immuno-modulatory fibrin-based biomaterials for use in bleeding and thrombosis, 3) probe the role of inflammation in the regulation of hemostasis and thrombosis, 3) explore mechanisms for modulating the mechanical and structural properties of fibrin, and 4) develop novel immune-mediated treatments for thrombosis.

Education: PhD Biomedical Engineering. June 2017. Drexel University. MS Biomedical Engineering. June 2013. Drexel University. BS Biomedical Engineering. June 2013. Drexel University.

Research/Work Experience: 07/2009-09/2009 STAR Research Scholar, Drexel University 03/2010-06/2013 Co-op Researcher, Children’s Hospital of Philadelphia 07/2013-06/2016 National Institute of Health Graduate Student Trainee, University of Pennsylvania 07/2016-06/2017 American Heart Association Graduate Research Fellow, University of Pennsylvania 07/2017-Present National Institute of Health Postdoctoral Fellow, University of Pennsylvania

Selected Publications: V. Tutwiler, A.D. Peshkova, G. Le Mihn, S. Zaytsev, R.I. Litvinov, D.B. Cines, J.W. Weisel. Blood clot contraction differently modulated internal and external fibrinolysis. Journal of Thrombosis and Hemostasis. 2019 17(2):361-370. V. Tutwiler, A.R. Mukhitov, A.D. Peshkova, G. Le Minh, R.R. Khismatullin, J. Vicksman, C. Nagaswami, R.I. Litvinov, J.W. Weisel Shape changes of erythrocytes during blood clot contraction and the structure of polyhedrocytes. Scientific Reports. 2018 8:17907 V. Tutwiler, H. Wang, R.I. Litvinov, J.W. Weisel. V.B. Shenoy. Interplay of Platelet Contractility and Elasticity of Fibrin/Erythrocytes in Blood Clot Contraction. Biophys J. 2017. 2017;112(4):714-723. Featured on cover. AD Peshkova, LM Giang, V Tutwiler, I.A. Andrianova, J.W. Weisel, R.I. Litvinov. A new link between inflammation and blood clotting: activated monocytes enhance clot contraction. Scientific Reports. 2017;7:5149. V. Tutwiler, A.D. Peshkova, I.A. Andrianova, D.R. Khasanova, J.W. Weisel, R.I. Litvinov. Contraction of blood clots is impaired in acute ischemic stroke. Athero Thromb Vasc Biol. 2017;37:271-279. Featured on cover. V. Tutwiler, R.I. Litvinov, A.P Lozhkin, A.D. Peshkova, T. Lebedeva, F.I. Ataullakhanov, K.L. Spiller, D.B. Cines, J.W. Weisel. Kinetics and mechanics of clot contraction are governed by the molecular and cellular composition of the blood. Blood. 2016;127(1):149-159. V. Tutwiler, D. Madeeva, H. Ahn, I. Andrianova, V. Hayes, X.L. Zheng, C.B. Cines, S. McKenzie, M. Poncz, L. Rauova, L. Platelet transactivation by monocytes promotes thrombosis in heparin-induced thrombocytopenia. Blood. 2016;127(4):464-472. Featured on cover.

Awards/Honors: Grant Funding: 1) NIH K99 Pathway to Independence (approved for funding). 2) NIH T32 Postdoctoral Training Grant, July 2017 –Present. 3) American Heart Association Predoctoral Fellow, July 2016-June 2017. 4) NIH T32 Predoctoral Training Grant, 2013-2016. 5) American Society of Hematology Trainee Grant, 2010-2013. Research Awards: 1) Finalist, Regeneron Prize for Creative Innovation, April 2019. 2) Travel to a mentor award, Thrombosis and Hemostasis Societies of North America, October 2018. 3) Best Science Image, Cell and Developmental Biology Retreat, May 2018. 4) Outstanding Dissertation Award, School of Biomedical Engineering, Sciences and Health Systems June 2017. 5) Best of the American Society of Hematology Annual Conference, 2012 Leadership Awards: James Herbert Outstanding Leadership Award, Drexel University, June 2017 Abstract Achievement/Travel Awards: ASCB – 2018. Fibrinogen Workshop – 2018. ASH – 2011,2012,2014,2018. SEBASTIEN G.M. UZEL, PhD Harvard University, 58 Oxford street, ESL 101R, Cambridge, Massachusetts, 02138 . [email protected]

Research Overview: De novo manufacturing of functional living tissues, as disease models, drug screening platforms, or for therapeutic applications has been one of the greatest challenges in the field of tissue engineering. While progress is being made in generating patient-specific functional cells, and assembling them around a vascular system to reach centimeter-scale constructs, approaching in vivo-like function will require sensing, processing, and control through a neural interface, especially in excitable organs such as the heart or skeletal muscles. My long-term interest is to develop the tools to understand and guide the formation and remodeling of synapses of the sensory-somatic or autonomic systems and inform the manufacturing of bionic functional tissues. Currently, in Prof. Lewis’ lab, at the crossroads between tissue engineering and materials science, my efforts focus on developing 3D printing methods and understanding assembly principles in order to vascularize patient-specific functional cardiac tissues towards therapeutic applications. Starting with iPSC-derived contractile building blocks, we demonstrated that once compacted, those cell aggregates exhibited the shear-thinning and viscoplastic rheological properties necessary to serve as a living matrix in which a sacrificial material could be free-form embedded printed into a hierarchical vascular template. Once evacuated, those channels could be endothelialized and perfused to maintain the viability of a tissue construct comprising nearly a billion cells. Prior to that experience, as a graduate student in Prof. Kamm’s and Prof. So’s labs at MIT, I designed microfluidic 3D culture systems to investigate the formation and function of neuromuscular junctions. Using optogenetic tools to interrogate the synapses and microfabrication to offer a physiologically-relevant microenvironment, we generated a study platform that helped us understand the pathophysiology of motor neuron diseases such as ALS. With these past experiences in tissue manufacturing via 3D bioprinting, biomaterials synthesis and characterization, microfluidic design and microfabrication, genetic and optogenetic engineering, combined with an undergrad training in modeling and computational biomechanics, I plan to apply a multidisciplinary approach to elucidate and guide the formation of biohybrid instrumented muscular tissues and provide a stimulating environment for my future trainees

Education: Ph.D. 2015 Department of Mechanical Engineering Massachusetts Institute of Technology, Cambridge, MA. M.S. 2007 Engineering Sciences Ecole Centrale Paris (ECP), Paris. M.S. 2007 Biomechanics Ecole Nationale Superieure d'Arts et Metiers (ENSAM), Paris.

Research/Work Experience: 2015-present Postdoctoral Fellow - Harvard University. Advisor: Jennifer A. Lewis. 2010-2015 Graduate research assistant - MIT. Advisors: Roger D. Kamm and Peter T.C. So 2008 Research engineer - National veterinary school of Alfort - Maisons-Alfort, Paris, France.

Selected Publications: 1. Skylar-Scott MA*, Uzel SGM*, Nam LL, Ahrens J, Truby RL, Damaraju S & Lewis JA. (2019). Biomanufacturing of organoid based tissues with embedded vasculature. Science Advances, in press. 2. Osaki T, Uzel SGM, & Kamm RD. (2019). Microphysiological 3D model of amyotrophic lateral sclerosis (ALS) from human iPSderived muscle cells and optogenetic motor neurons. Science Advances, 4(10), eaat5847. 3. Uzel SGM, Platt RJ, Subramanian V, Pearl TM, Rowland CJ, Chan V, So PTC, & Kamm RD. (2016). Microfluidic device for the formation of optically excitable, three-dimensional, compartmentalized motor units. Science Advances, 2(8), e1501429. 4. Uzel SGM, Amadi OC, Pearl TM, Lee RT, So PTC, & Kamm RD. (2016). Simultaneous or sequential orthogonal gradient formation in a 3D cell culture microfluidic platform. Small, 12(5), 688. 5. Uzel SGM, Pavesi A, & Kamm RD. (2014). Microfabrication and microfluidics for muscle tissue models. Progress in Biophysics and Molecular Biology, 115(2), 279–293. 6. Polacheck WJ*, Li R*, Uzel SGM*, & Kamm RD. (2013). Microfluidic platforms for mechanobiology. Lab on a Chip, 13(12), 2252–2267.

Awards/Honors: 2013 Outstanding Paper Award - 2013 ASME Global Congress on NanoEngineering for Medicine and Biology (NEMB). 2012 & 2013 Awardee of the Jean Gaillard Memorial Fellowship. 2010 Laureate of the Robert B. Guenassia Award. 2008-2009 Recipient of the MIT Presidential Fellowship. MAX MANUEL VILLA, PhD Molecular Genetics and Microbiology, Duke University, 917 Vickie Drive, Cary, North Carolina, 27511 . [email protected]

Research Overview: The human microbiome encodes a vast 'second genome' that dwarfs our own, and plays critical roles in human health, development, and disease. For instance, microbes in our guts modulate the immune system, provide protection from pathogen colonization, and allow us to extract more energy from our diet. Furthermore, gut microbes have been leveraged therapeutically in the treatment of antibiotic resistant C. difficile and can modify pharmaceutical compounds, tightly linking gut microbes to how we already treat disease. Yet the mechanistic insight required for next generation therapies is lacking in part since the tools for microbial culture were developed hundreds of years prior and the diversity of the human microbiome is staggeringly vast. I have developed new tools for dealing with the incredible diversity of the microbiome that examine function with strain level resolution. My research will use and build on these tools to investigate how the microbiome shapes human health and disease outcomes, and ultimately realize new microbiome targeted therapeutics.

Education: Ph.D. Materials Science & Engineering, 2014, University of Connecticut M.S. Mechanical Engineering, 2009, University of Connecticut B.S. Mechanical Engineering, 2007, University of Connecticut

Research/Work Experience: ·Burroughs Wellcome PDEP Fellow, 2015-Present, Center for Genomic and Computational Biology, Department of Molecular Genetics and Microbiology, Duke University, Durham, NC ·GAANN Predoctoral Fellow, 2010-2014, Center for Regenerative Medicine and Skeletal Development, University of Connecticut, Connecticut Health Center, Farmington, CT and Department of Materials Science and Engineering, University of Connecticut, Storrs, CT ·Associate Scientist, 2009-2010, Surgical Devices R&D, Covidien, North Haven, CT ·Graduate Student, 2007-2009 Department of Mechanical Engineering, University of Connecticut, Storrs, CT

Selected Publications: Villa M.M., Bloom, R.J., Silverman, J.D., Durand, H.K., Jiang, S., Wu, A., Huang, S., You, L., David, L.A., High-throughput isolation and culture of human gut bacteria with droplet microfluidics. (Submitted) Villa M.M., Bloom, R.J., David, L.A., RAPID PHENOTYPING AND IDENTIFICATION OF MICROBES FROM A COMPLEX MICROBIAL COMMUNITY. US Patent Number: 62/628,170, Filed: 02/08/2018, (patent pending) Villa M.M., Wang, L., Huang, J.P., Rowe, D.W., Wei, M., Bone tissue engineering with a collagen- hydroxyapatite scaffold and mouse bone marrow stromal cells in a critical size calvarial defect, Journal of Biomedical Materials Research Part B: Applied Biomaterials 103, 243-253 (2015). (Editor’s Choice) Villa M.M., Wang, L., Huang, J.P., Rowe, D.W., Wei, M., Visualizing Osteogenesis In Vivo within a Cell- Scaffold Construct for Bone Tissue Engineering using 2-Photon Microscopy, Tissue Engineering: Part C 19, 839-849 (2013). (cover image)

Awards/Honors: Burroughs Wellcome Fund Postdoctoral Enrichment Program, 2016-2019 ($60,000) NIH Research Supplement to Promote Diversity in Health-Related Research, Project title: Optimization of cell delivery to 3D scaffolds for in vivo osteogenesis, 2013-2014 ($43,000) Department of Education Graduate Assistantship in Areas of National Need (GAANN) Predoctoral Fellowship: Biomaterials for Tissue Regeneration, 2010-2013 ($60,000) HUA WANG, PhD John Paulson School of Engineering and Applied Sciences, Harvard University, 58 Oxford St, ESL 415, Cambridge, MA, 02138 . [email protected] Research Overview: One of my general research interests is to understand how cancer and immune cells can be manipulated and engineered to facilitate targeted delivery of therapeutics, in order to improve and innovate therapies for cancers, injured tissues, autoimmune disorders, and other diseases. For example, cancer cells can actively metabolize unnatural monosaccharides bearing chemical tags and express them in the form of glycoproteins. These cell-surface chemical tags (e.g., azide) enable targeted conjugation of molecules of interest via efficient chemistries to monitor or treat cancer cells. I have been interested in exploring selective labeling of cancer cells in vivo, for subsequent development of cancer-targeted therapies. Similarly, unnatural sugars can also metabolically label immune cells with chemical tags, which will be utilized for targeted modulation of dendritic cells (DCs) and T cells with immunomodulatory agents in my laboratory, in the context of cancers and autoimmune disorders. Biomaterials loaded with chemokines provide a powerful tool to concentrate and manipulate immune cells, including DCs and T cells, in situ, and can be utilized to develop cancer therapies for cancers and injured tissues. In the context of cancers, I aim to develop biomaterial-based cancer vaccines for the treatment of poorly-immunogenic tumors, and explore biomaterials that can maintain the survival and proliferation of adoptively transferred T cells. Immune responses also play a critical role in tissue repair and implant integration, and I am particularly interested in deciphering the role of T cells in orchestrating tissue regeneration, and utilizing biomaterials to recruit and modulate T cells in situ to facilitate tissue restoration.

Education: 08/2012-06/2016, Ph.D., Materials Science and Engineering, University of Illinois at Urbana-Champaign 08/2008-06/2012, B.S., Polymer Science and Engineering, University of Science and Technology of China

Research/Work Experience: 07/2017-Present Wyss Technology Development Fellow 07/2016-Present Postdoctoral Fellow, John A. Paulson School of Engineering and Applied Sciences, Wyss Institute for Biologically Inspired Engineering, Harvard University (Advisor: David J. Mooney)

Selected Publications: 1. Wang, H., Sobral, M., Cartwright, A., Zhang, D., Li, A., Dellacherie, M., Koshy, S., Wucherpfennig, K., Mooney, D. In Vivo Metabolic Labeling and Targeted Modulation of Dendritic Cells. Submitted to Nature Materials, 2019 (under review). 2. Wang, H., Sobral, M., Snyder, T., Brudno, Y., Gorantla, V., Mooney, D. Clickable, Acid Labile Immunosuppressive Prodrugs for In Vivo Targeting of Reloadable Drug Delivery Depots in Organ Transplantation. Submitted to Biomaterials, 2019 (under review). 3. Wang, H., Bo, Y., Liu, Y., Xu, M., Cai, K., Wang, R., Cheng, J. In Vivo Cancer Targeting via Glycopolyester Nanoparticle Mediated Metabolic Cell Labeling Followed by Click Reaction. Biomaterials 2019, 218, 119305. 4. Wang, H., Liu, Y., Xu, M., Cheng, J. Azido-Galactose Outperforms Azido-Mannose for Metabolic Labeling and Targeting of Hepatocellular Carcinoma. Biomaterials Science 2019. DOI: 10.1039/C9BM00898E. 5. Wang, H., Najibi, A., Sobral, M., Lee, J., Li, A., Verbeke, C., Mooney, D. Antigen-Free Cancer Vaccine to Treat Poorly Immunogenic Tumors. Nature Communication, 2019. Under revision. 6. Wang, H., Mooney, D. Biomaterial-Assisted Targeted Modulation of Immune Cells in Cancer Treatment. Nature Materials 2018, 17, 761-772. 7. Wang, H., Wang, R., Cai K., He H., Liu, Y., Yen, J., Wang, Z., Xu, M., Sun, Y., Zhou, X., Yin, Q., Tang, L., Dobrucka, I.T., Dobrucki, L.W., Chaney, E.J., Boppart, S.A., Fan, T.M., Lezmi, S., Chen, X., Yin, L., Cheng, J. Selective In Vivo Cell Labeling Mediated Cancer Targeting. Nature Chemical Biology 2017, 13, 415-424. 8. Wang, H., Tang, L., Liu, Y., Dobrucka, I.T., Dobrucki, L.W., Yin, L., Cheng, J. In Vivo Targeting of Metabolically Labeled Cancers with Ultra-Small Silica Nanoconjugates. Theranostics 2016, 6, 1467-1476. 9. Wang, H., Gauthier, M., Kelly, J. R., Miller, R.J., Xu, M., O’Brien, W.D., Cheng, J. Targeted Ultrasound Assisted Cancer- Selective Labeling and Imaging. Angewandte Chemie International Edition 2016, 55, 5452-5456. 10. Wang, H., Xu, M., Xiong, M., Cheng, J. Reduction-Responsive Dithiomaleimide-Based Nanomedicine with High Drug Loading and FRET-Indicated Drug Release. Chemical Communications 2015, 51, 4807-4810. 11. Wang, H., Tang, L., Tu, C., Song, Z., Yin, Q., Yin, L., Zhang, Z., Cheng, J. Redox-Responsive, Core-Cross-Linked Micelles Capable of On-Demand, Concurrent Drug Release and Structure Disassembly. Biomacromolecules 2013, 14, 3706- 3712. HUILIANG WANG, PhD Bioengineering, Stanford University, W250 Clark Center, 318 campus drive, Stanford, CA, 94305 - [email protected]

Research Overview: Recent developments in genetics, materials, and electronics have enabled a deeper understanding of the nervous system and enhanced therapeutic solutions in neural diseases. For example, current developments in optogenetics have demonstrated that modulating the neural activity of specific projections in the brain is capable of rescuing complex behavioral deficits, including those with implications for depression, autism, and anxiety. However, this approach requires the genetic modification of neurons via viral transduction of visible light transducers, and light penetration depth is limited by the scattering of visible light. During my postdoctoral research with Karl Deisseroth, I have developed a method to achieve non-genetic, projection-specific neural modulation in freely behaving mice. Specifically, I found that suitably functionalized gold nanorods are rapidly internalized by neuron soma or axon terminals, are transported retrogradely/anterogradely through axons and are sufficient to modulate neural activity under near-infrared light after axonal transport both in vitro and in vivo (this work is currently funded by a NIH K01 award). Furthermore, I completed my graduate training with Zhenan Bao and developed better carbon nanotubes/polymers based flexible/wearable electronics, with applications in areas such as health monitoring and neuromodulation. Combining my PhD background in materials processing and device fabrication with my postdoctoral training in neurotechnology and animal behaviors, I plan to establish my future faculty program in developing novel nanomaterials and minimal invasive electronic devices for targeted modulation and recording of neural activity.

Education: 2010-2014: PhD in Materials Science and Engineering, Stanford University, 2006-2010: BEng/MEng in Material Science, University of Oxford

Research/Work Experience: 2015-Present: Postdoctoral Scholar, Department of Bioengineering, Stanford University (Advisor: Karl Deisseroth) 2010-2014: PhD Student, Department of Materials Science and Engineering, Stanford University (Advisor: Zhenan Bao) 2009-2010: Master Student, Department of Materials Science, University of Oxford (Advisor: Jamie Warner and Andrew Briggs)

Selected Publications: -H. Wang, L. Fenno, Y. Chen, C. Kim, C. Ramakrishnan, M. Inoue, S.Gambhir, K.Deisseroth, Projection-Specific Modulation of Neural Activity with a Non-Genetic Method, In preparation -H. Wang, Y. Wang, B.T.C. Keong, K. Kim, J. Lopez, C. Wei, Z. Bao, Shape-Controlled, Self-Wrapped Carbon Nanotube Electronics, Advanced Science, 2, 1500103, (2015) -H. Wang, K. N. Houk, Z. Bao et al., N-type Conjugated Polymer-Enabled Selective Dispersion of Semiconducting Carbon Nanotubes for Flexible CMOS-like Circuits. Advanced Functional Materials, 25, 1837-1844 (2015) -H. Wang, B. Cobb, A. Breemen, G. Gelinck, Z. Bao, Highly Stable Carbon Nanotube Top-Gate Transistors with Tunable Threshold Voltage, Advanced Materials, 26, 4588-4593 (2014) -H. Wang, Y.Cui, Z. Bao et al., Tuning the Threshold Voltage of Carbon Nanotube Transistors by n-Type Molecular Doping for Robust and Flexible Complementary Circuits, Proceedings of the National Academy of Science (PNAS), 111(13), 4776-4781 (2014) -H. Wang, K. Houk, Z. Bao et al., High-Yield Sorting of Small-Diameter Carbon Nanotubes for Solar Cells and Transistors, ACS Nano, 8, 2609-2617 (2014) -H. Wang, M.F.Toney, Z. Bao et al., Scalable and Selective Dispersion of Semiconducting Arc-Discharged Carbon Nanotubes by Dithiafulvalene/Thiophene Copolymers for Thin Film Transistors. ACS Nano, 26, 2659-2668 (2013)

Awards/Honors: 2018-2022: NIH Mentored Research Scientist Development Award (K01) 2016-2018: NIH National Research Service Award (NRSA) Postdoctoral Fellowship (F32) 2014: Chinese Government Award For Outstanding Self-Financed Students Abroad 2014: Gold Graduate Student Award, Materials Research Society (MRS) 2014: Graduate Research Award, American Vacuum Society (AVS) 2014: Excellence in Graduate Polymer Research Award, American Chemical Society (ACS) 2012-2014: Link foundation energy fellowship (3 winners awarded nationally in US each year) 2012: Best Research Poster at 2012 Global Climate & Energy Project (GCEP) Research Symposium 2012: Selected for “Overseas Talents in Chinese Academy of Science" activity week CHIA-LUNG WU, PhD Orthopaedic Surgery, Washington University in Saint Louis, One Brookings Drive, Saint Louis, MO, 63130 [email protected]

Research Overview: Human pluripotent stem cells (hPSCs) provide a promising cell source for tissue engineering and cell therapy, as well as in vitro disease modeling and drug screening for many musculoskeletal disorders. However, how to efficiently differentiate hPSCs into specific musculoskeletal tissues remains challenging. Thus, my research program will focus on (1) investigating gene regulatory networks (GRNs) and epigenetic mechanisms governing cell fate decisions toward musculoskeletal lineages and (2) leveraging newly identified genetic and/or epigenetic targets to enhance therapeutic applications of stem cells and tissue engineering for musculoskeletal diseases. My goal is to use interdisciplinary approaches including sequencing technology, bioinformatics, synthetic biology, biomaterials, and animal models to examine mechanistic hypotheses within this overarching theme. During my doctoral research in Dr. Farshid Guilak′s lab, I investigated links between obesity and stem cell function using DNA methylation sequencing and transgenic mouse models. Our finding of hypomethylated status in obese stem cells versus lean stem cells provides a plausible mechanism for dysregulated stem cell multipotency in obesity. For my postdoctoral training, I studied embryonic cartilage development using hPSCs as a model system. Utilizing a CRISPR-Cas9 edited reporter hPSC line, I established a robust step-wise chondrogenic differentiation protocol for hPSCs. I have further expanded my expertise in the single-cell RNA sequencing (scRNA-seq) and bioinformatics. Through re-constructing GRNs from scRNA-seq data of hPSC chondrogenesis, I identified hub genes governing off-target differentiation and significantly eliminated undesired cell lineages by targeting their molecular pathways. I also demonstrated that tissue-engineered cartilage constructs derived from hPSCs can be used to repair osteochondral defect in mice in vivo. Additionally, I have uncovered several epigenetic regulators putatively involved in cell fate specification during chondrogenesis. I was recently awarded NIH K99/R00 to dissect molecular mechanisms of these epigenetic regulators using chromatin immunoprecipitation followed by sequencing (ChIP-seq) techniques and tissue-specific knockout mouse models. Collectively, my training has built a foundation that allows me to apply multiple disciplinary approaches to investigate the molecular mechanism regulating the development and homeostasis of musculoskeletal tissues, with the goal of identifying novel therapeutic targets for promoting tissue engineering and regenerative medicine.

Education: · Ph.D., Biomedical Engineering, Duke University, 2015 · M.S., Material Science and Engineering, National Taiwan University, 2005 · B.S., Materials and Mineral Resources Engineering, National Taipei University of Technology, 2001

Research/Work Experience: · Post-doctoral research associate, Washington University in Saint Louis, 2015-present · Graduate Research, Duke University, 2009-2015 · Teaching Assistant (full time), Material Science and Engineering, National Taiwan University, 2005-2007 · Operation Corporal, Defense Command Post, Dept. of Defense, Taiwan, 2001-2003

Selected Publications: 1. Wu CL*, Dicks A*, Steward N, et al., ″Single Cell Transcriptomic Analysis of Human Pluripotent Stem Cell Chondrogenesis″, (*co-first author; under review, Cell Stem Cell). 2. Wu CL*, Dicks A*, Steward N, et al., ″Prospective Isolation of Chondroprogenitors from Human iPSCs Based on Cell Surface Markers Identified using a CRISPR-Cas9-Generated Reporter″, (in revision Stem Cell Res Ther). 3. Adkar SS*, Wu CL*, et al., ″Step-Wise Chondrogenesis of Human Induced Pluripotent Stem Cells and Purification Via a Reporter Allele Generated by CRISPR-Cas9 Genome Editing″, Stem Cells. 2019;37(1):65-76. 4. Wu CL, McNeill J, Goon K, et al., ″Conditional Macrophage Depletion Increases Inflammation and Does Not Inhibit the Development of Osteoarthritis in Obese Macrophage Fas-Induced Apoptosis Transgenic Mice″, Arthritis Rheum. 2017;69(9):1772-83. 5. Wu CL, Jain D, McNeill JN, et al., ″Dietary Fatty Acid Content Regulates Wound Repair and the Pathogenesis of Osteoarthritis Following Joint Injury″, Ann Rheum Dis. 2015;74(11):2076-83.

Awards/Honors: · NIH NIAMS K99/R00 Pathway to Independence Award, 1K99AR075899, July 2019-present. · The New Investigator Recognition Awards (NIRA), Orthopaedic Research Society (ORS), 2018 & 2014. · Studying Abroad Scholarship, Ministry of Education, Taiwan, ($32,000 total), 2012

MENGI WU, PhD University of Michigan, 2644 GG Brown, Ann Arbor, Michigan, 48105. [email protected]

Research Overview: My research is focused on developing novel tools for disease diagnosis and precision medicine using micro/nano technology. Integrating acoustics and microfluidics, I have developed a series of acoustofluidic technologies that are able to separate and enrich various biomarkers, including exosomes, circulating tumor cells and blood components, for disease early-diagnosis and personlized treatment. Due to the advantages of high biocompatibility, ease of operation and controllability, the acoustofluidic technologies I developed are of great potential for liquid biopsy in biomedical and clinical applications. Using MEMS technology, the miniaturized gas chromatography system has developed for rapid and handheld system for the detection and analysis of volatile organic compounds (VOC). With efforts on further integration of individual components, I am aiming to turn the microGC systems to be portable, and multi-dimensional for bedside VOC biomarker analysis and breath biopsy. My research were highlighted by National Science Foundation (three times) and reported by over 300 public media. Four of my papers were ranked top 5% of all research outputs compared to outputs of the same age.

Education: 2014-2018, Pennsylvania State University, Ph.D. in Engineering Sciences and Mechanics, Advisor: Professor Tony Jun Huang 2010-2013, Peking University, M.S. in Micro Electro Mechanical Systems, Advisor: Prof. Zhihong Li 2006-2010, Peking University, China, B.S. in Microelectronics

Research/Work Experience: •September 2018 – present, Postdoctoral fellow, Department of Mechanical Engineering, University of Michigan •August 2016 – August 2018, Associate in Research, Department of Mechanical Engineering and Materials Science, Duke University. •August 2014 - August 2018, Research Assistant, Engineering Sciences and Mechanics, Pennsylvania State University. •July 2013 – July 2014, Research & Development Engineer, Etta Biotech Co. Ltd, Suzhou, China.

Selected Publications: •Wu, Mengxi, et al. "Acoustofluidic separation of cells and particles." Microsystems & nanoengineering 5.1 (2019): 32. •Wu, Mengxi, et al. "Isolation of exosomes from whole blood by integrating acoustics and microfluidics." Proceedings of the National Academy of Sciences 114.40 (2017): 10584-10589. •Wu, Mengxi, et al. "Circulating Tumor Cell Phenotyping via High‐Throughput Acoustic Separation." Small 14.32 (2018): 1801131. •Wu, Mengxi, et al. "Acoustic separation of nanoparticles in continuous flow." Advanced functional materials 27.14 (2017): 1606039. •Wu, Mengxi, et al. "Separating extracellular vesicles and lipoproteins via acoustofluidics." Lab Chip 19.7 (2019): 1174-1182. •Wu, Mengxi, et al. "High-throughput cell focusing and separation via acoustofluidic tweezers." Lab Chip 18.19 (2018): 3003-3010. •Wu, Mengxi, et al. "Method for electric parametric characterization and optimization of electroporation on a chip." Analytical chemistry 85.9 (2013): 4483-4491. •Wu, Mengxi, et al. "High-density distributed electrode network, a multi-functional electroporation method for delivery of molecules of different sizes." Scientific reports 3 (2013): 3370.

Awards/Honors: •Thomas and June Beaver Fund Award, 2018, Pennsylvania State University •College of Engineering Graduate Excellence Fellowship, 2014 - 2017, Pennsylvania State University YAOYING WU, PhD Biomedical Engineering, Duke University, 101 Science Dr, Durham, North Carolina, 27708 [email protected] Research Overview: My independent research will seek to advance our understanding of the interactions between biomaterials and the immune system through the design of polymeric platforms to 1) deliver antigens through receptor targeting for enhanced immune responses; 2) modulate local immune microenvironments for immune cell polarization and education, and; 3) improve antibody maturation by engaging cellular and chemical cues. During my doctoral training with Dr. Theresa Reineke, I developed a series of glucose-derived cationic glycopolymers that form very stable complexes with plasmid DNA in the presence of serum. These glycopolymers also specifically engage asialoglycoprotein receptors on hepatocytes, leading to elevated cellular uptake and transfection efficiency. Seeking to better understand the immunological aspects of biomaterials, I joined Dr. Joel Collier’s lab as a postdoctoral scholar and developed a novel α-helical self-assembling nanofiber vaccine delivery platform (Coil29). The self- assembled nanofibers can elicit strong humoral and cellular immune responses without provoking significant inflammatory reactions. I also discovered that T cell epitopes within the primary Coil29 peptide sequence drive T follicular helper cell differentiation, leading to its superior ability to stimulate humoral responses relative to conventional adjuvants. We were awarded an international patent for this technology. My current efforts are focused on leveraging the multivalency and strong immunogenicity of this platform to develop an effective combinatory cancer vaccine therapy. My doctoral training in polymer design, combined with the immunological expertise gained from my postdoctoral research, makes me uniquely qualified to carry out my proposed research. Through the engagement of defined immunological pathways, these proposed platforms will fill the growing clinical need for targeted therapies to a range of conditions, from infectious diseases to cancer.

Education: • PhD, Chemistry, University of Minnesota, Twin Cities, 2014 • M.S. Polymer Chemistry and Physics, Beijing University of Chemical Technology, 2009 • B.E. Materials Science and Engineering, Tianjin University, 2006

Research/Work Experience: • 2016-present Postdoctoral Scholar, Duke University, Advisor: Joel Collier, PhD • 2014-2016, Postdoctoral Scholar, University of Chicago, Advisor: Joel Collier, PhD • 2011-2014, Graduate Research Assistant, University of Minnesota, Twin Cities, Advisor: Theresa Reineke, PhD • 2009-2011, Graduate Research Assistant, Virginia Tech, Advisor: Theresa Reineke, PhD • 2006-2009, Graduate Research Assistant, Beijing University of Chemical Technology, Advisor: Zhifeng Fu, PhD

Selected Publications: • Nelson CE, Wu Y, Gemberling MP, Oliver ML, Waller MA, Bohning JD, Robinson-Hamm JN, Bulaklak K, Castellanos Rivera RM, Collier JH, Asokan A, Gersbach CA, Long-term evaluation of AAV-CRISPR genome editing for duchenne muscular dystrophy, Nature Medicine 2019, 25(3), 427 • Wu Y, Norberg PK, Reap EA, Congdon K, Fries C, Kelly SH, Sampson JH, Conticello VP, Collier JH, A supramolecular vaccine platform based on α-helical peptide nanofibers, ACS Biomaterials Science & Engineering 2017, 3(12), 3128 • Wu Y, Smith AE, Reineke TM, Lipophilic polycation vehicles display high plasmid DNA delivery to multiple cell types, Bioconjugate Chemistry 2017, 28(8), 2035 • Wu Y, Collier JH, α-Helical coiled coil peptide materials for biomedical applications, WIREs Nanomedicine and Nanobiotechnology 2017, 9(2), e1424 • Wu Y, Wang M, Sprouse D, Smith AE, Reineke TM, Glucose-containing diblock polycations exhibit molecular weight, charge, and cell-type dependence for pDNA delivery, Biomacrolecules 2014, 15(5), 1716 • Sizovs A, Xue L, Tolstyka ZP, Ingle NP, Wu Y, Cortez MA, Reineke TM, Poly(trehalose): sugar-coated nanocomplexes promote stabilization and effective polyplex-mediated siRNA delivery, Journal of the American Chemical Society 2013, 135(41), 15417

Awards/Honors: • 2019, Duke Incubation Fund (Co-Principle Investigator) • 2018, Duke Biomedical Engineering Kewaunee Poster Award • 2014, American Chemical Society Excellence in Graduate Polymer Research Award • 2014, The Center for Genome Engineering of University of Minnesota Travel Award • 2014, American Chemical Society Graduate Student Travel Grant ALEXANDER M. XU, PhD Institute for Systems Biology, 401 N Terry Ave, Seattle, WA, 98109 [email protected] Research Overview: My research lab will develop new methods to study single cells in multiple dimensions, to better understand cell heterogeneity and cancer biology. We will focus on combining transcriptomics with phenotypic and phenomenological measurements to pair transcriptomic analysis and bioinformatic inference with functional ground truths. To make multiple orthogonal measurements in a single cell (fluorescence microscopy + transcriptomics for instance), we will develop new devices and materials, chemistries, and bioengineering strategies. We will then use systems biology and computational approaches to process these multi-dimensional data sets, and study the heterogeneity of cancer phenomena. We will target questions such as cell invasiveness, metabolism, and adhesion, that are largely invisible to the primary –omics methods, but have great clinical significance. We will seek out collaborations to expand single cell analysis to partners across biomedical engineering topics, especially labs that would benefit from integrating singlecell transcriptomics into a flagship assay or measurement.

Education: Stanford University, Palo Alto, CA Ph.D in Materials Science and Engineering, 2015 Massachusetts Institute of Technology, Cambridge, MA B.S. in Materials Science and Engineering, 2010 B.S. in Mathematics, 2010

Research/Work Experience: Institute for Systems Biology, Since 2018 Postdoctoral Fellow – James Heath California Institute of Technology, 2015-2018 Postdoctoral Fellow – James Heath Stanford University, 2010-2015 Graduate Student – Nicholas Melosh Harvard University, 2010 Research Intern – Irene Chen Massachusetts Institute of Technology, 2008-2010 Undergraduate Researcher – Angela Belcher Massachusetts Institute of Technology, 2007-2010 Undergraduate Researcher – Forest White Argonne National Laboratory, 2008-2009 Research Intern – Jeffrey Elam

Selected Publications: 1. Alexander M Xu, Q Liu, K Takata, S Jeoung, Y Su, I Antoshechkin, S Chen, M Thomson, JR Heath. Integrated Measurement of Cytosolic Proteins and Transcripts in Single Cells. Lab on a Chip. (2018) -Front cover 2. *J Sander, *SV Schmidt, B Cirovic, N Mcgovern, O Papontanopoulou, A-L Hardt, AC Aschenbrenner, C Kreer, T Quast.,Alexander M Xu, LM Schmidleithner,… NA Melosh, EW Nowell, F Ginhoux, A Schlitzer, JL Schultze. of Human Monocytes Is Regulated by Time-Dependent Interleukin-4 Signaling and the Transcriptional Regulator NCOR2. Immunity. (2017) 3. Alexander M Xu, A Aalipour, S Leal-Ortiz, AH Mekhdjian, X Xie, AR Dunn, CC Garner, NA Melosh. Quantification of Nanowire Penetration into Living Cells. Nature Communications. (2014) 4. Alexander M Xu*, JJ VanDersarl*, NA Melosh. Nanostraws for Direct Fluidic Intracellular Access. Nano Letters. (2011) -Front cover 5. Alexander M Xu and PH Huang. Receptor Tyrosine Kinase Coactivation Networks in Cancer. Cancer Research. (2010)

Awards/Honors: NIH F32 Ruth Kirschstein Postdoctoral Fellowship, 2017 NSF Graduate Research Fellow, 2010 NDSEG Fellowship, 2010 JIANGSHENG XU PhD Fischell Department of Bioengineering, UNIVERSITY OF MARYLAND, COLLEGE PARK, 426 ridge rd apt12, Greenbelt, MD, 20770 . [email protected]

Research Overview: I have keen interest in biomedical research with a broad background in materials science, chemistry, cancer biology, bioinformatic and biomedical engineering, with specific training and experts in cancer detection, nanomedicine, mitochondria targeting, and target gene therapy. I have more than 9 years of experience in cancer research on developing of novel nanomaterial-based therapeutic strategies, e.g. targeting triple negative breast cancer (TNBC) with hemizygous TP53 deletion. My career goal is to develop an independent research program in cancer research at the forefront of emerging engineering technologies and biological knowledge guided by understanding of clinical needs. My overarching objectives as an investigator in an academic environment institution focus on developing targeted therapeutic strategies that leverage and combine promising research from the fields of biomaterials, cancer biology, bioinformatics, and biomedical engineering. The topics will across a broad spectrum of cancer research, spanning fundamental, basic preclinical, and translational work, which covers cancer research offering new insights into cancer biology, genetics and bioinformatics, new approaches for the development and delivery of diagnostics and therapies, and new ways of evaluating drugs of cancer. This applications include but not limited to: 1. To explore the POLR2A-targeted therapeutic strategy for cancers harboring TP53 deletion, such as PDAC 2. To build a 3D PDAC tumor model for understanding and evaluating the POLR2A-targeted therapeutic strategy 3. To develop artificial tumors that replicate the stromal, immune, extracellular matrix, and/or vascular composition in the tumor microenvironment at the molecular and cell level. 4. Microfluidics based one single cell encapsulation, developing, and analysis. 5. Machine learning associated therapeutic targets identification in human cancer.

Education: Ph.D. in Materials Science, 2010-2015, South China University of Technology B.S. in Polymer Science and Engineering, 2006-2010, Wuhan Institute of Technology

Research/Work Experience: Postdoctoral Research Associate, Advisor: Xiaoming He, University of Maryland, College Park MD, 12/2017- Postdoctoral Research Associate, Advisor: Xiaoming He, The Ohio State University, Columbus OH, 12/2015-11/2017 Graduate Research Fellow; Advisor: Fang Zeng, South China University of Technology, China, 09/2010-05/2015

Selected Publications: (1) Xu, J.; Liu, Y.; Li, Y.; Wang, H., Stewart, S.; Agarwal, P.; Zhang, Y.; Liu, S.; Zhao, G.; Wan, J.; Lu, X.; He, X. Precise Targeting of POLR2A as a Therapeutic Strategy for Human Triple Negative Breast Cancer. Nature Nanotechnology. 2019, 14, 388 (2) Liu, Y.*; Xu, J.*; Choi, H. H.*; Han, C.; Fang, Y.; Li, Y.; Van der Jeught, K.; Xu, H.; Zhang, L.; Frieden, M.; et al. Targeting 17q23 Amplicon to Overcome the Resistance to Anti-HER2 Therapy in HER2+ Breast Cancer. Nature Communications. 2018, 9 (1), 4718. (*equal contribution) (3) Xu, J.; Zeng, F.; Wu, H.; Yu, C.; Wu, S. Dual-Targeting Nanosystem for Enhancing Photodynamic Therapy Efficiency. ACS Appl. Mater. Interfaces 2015, 7 (17), 9287. (4) Xu, J.; Zeng, F.; Wu, H.; Hu, C.; Wu, S. Enhanced Photodynamic Efficiency Achieved via a Dual-Targeted Strategy Based on Photosensitizer/Micelle Structure. Biomacromolecules 2014, 15 (11), 4249. (5) Xu, J.; Zeng, F.; Wu, H.; Hu, C.; Yu, C.; Wu, S. Preparation of a Mitochondria-Targeted and NO-Releasing Nanoplatform and Its Enhanced pro-Apoptotic Effect on Cancer Cells. Small 2014, 10 (18), 3750.

Awards/Honors: Pelotonia Postdoctoral Fellowship, 06/2016-11/2017, The Ohio State University Comprehensive Cancer Center (OSUCCCC) University First-Class Pre-Doctoral Research Award, 06/2012-06/2015, South China University of Technology, China YUAN YANG, PhD 1Physical Therapy and Human Movement Sciences, Northwestern University, 645 N Michigan Ave, Suite 1100, Chicago, IL, 60611, 2Northwestern University, Chicago, IL, 60611 . [email protected] Research Overview: My research focuses on investigating functional neural connectivity in the sensorimotor system during normal and pathological motor control. It allows revealing how the sensorimotor system adapts and reorganizes after neurological disorders (e.g. stroke) and during the recovery, so as to evaluate motor impairments and facilitate science-driven neuro- rehabilitation interventions. My current research integrates biologically realistic computer simulations and quantitative experimental approaches based on EEG, EMG, T1 MRI, DTI, non-invasive brain stimulation (TMS/tDCS) and mechatronic devices. It aims to improve our understanding of neural mechanisms underlying abnormal functional connectivity as well as their link to motor impairments following a unilateral brain injury. Besides this main research line, I am also interested in 1) bio-system identification for understanding how the brain processes somatosensory inputs, and 2) EEG-based brain-computer interfaces for motor rehabilitation. My research is supported by NIH and Northwestern Memorial Foundation.

Education: B.S. in Biomedical Engineering, 2008, Central South University, China. M.S. in Biomedical Engineering, 2010, Shanghai Jiao Tong University, China. Ph.D. in Signal and Image Processing, 2013, Telecom ParisTech/CNRS, France.

Research/Work Experience: 2013/11-2017/07 Postdoc Researcher, Delft University of Technology, Department of Biomechanical Engineering, Delft, The Netherlands. 2017/03 Visiting Scientist, QIMR Berghofer Medical Research Institute, System Neuroscience Group, Brisbane, Australia. 2017/07-Present, Research Assistant Professor, Northwestern University Feinberg School of Medicine, Department of Physical Therapy and Human Movement Sciences, Chicago, Illinois. 2019/06-Present, Preceptor, Northwestern University Interdepartmental Neuroscience Graduate Program, Chicago, Illinois. 2019/07-Present, Ad hoc Graduate Faculty, Northwestern University Graduate School, Evanston, Illinois.

Selected Publications: Selected from 22 SCI journal publications 1. Yang Y, Dewald, J. P. A., van der Helm, F. C. T and Schouten, A. C (2018), Unveiling neural coupling within the sensorimotor system: directionality and nonlinearity. European Journal of Neuroscience, 48(7): 2407-2415. 2. Yang Y, Solis-Escalante T, Yao J, Daffertshofer A, Schouten A.C., and van der Helm F.C.T. (2016), A general approach for quantifying nonlinear connectivity in the nervous system based on phase coupling, International Journal of Neural Systems, 26 (1):1550031. 3. Yang Y, Solis-Escalante T, Yao J, van der Helm F.C.T, Dewald J.P.A and Schouten A.C (2016), Nonlinear Connectivity in the Human Stretch Reflex Assessed by Cross-frequency Phase Coupling, International Journal of Neural Systems, 26(8): 1650043. 4. Yang Y, Solis-Escalante T, van der Helm F.C.T, Schouten A.C (2016), A generalized coherence framework for detecting and characterizing nonlinear interactions in the nervous system, IEEE Transactions on Biomedical Engineering, 63(12): 2629-2637. 5. Yang Y, Solis-Escalante T, van de Ruit M, van der Helm F.C.T and Schouten A.C (2016), Nonlinear coupling between cortical oscillations and muscle activity during isotonic wrist flexion, Frontiers in Computational Neuroscience, 10:126. PMID: 27999537.

Awards/Honors: 1. NIH/NICHD 1R21HD099710 2. Dixon Translational Research Award 3. Hojjat Adeli Award for Outstanding Contributions in Neural Systems, World Scientific Publishing (2017). MURAT YILDIRIM, PhD Picower Institute for Learning and Memory, Massachusetts Institute of Technology, 43 Vassar Street, Cambridge, MA, 02139 Email: [email protected], Website: https://yildirimm1983.wixsite.com/mysite

Research Overview: My research at MIT and UT Austin focuses on developing table-top and endoscopic multiphoton microscopy systems to better understand the structure and function of biological systems such as vocal folds and the brain. My work has an impact on both diagnosis and treatment of several disorders spanning from vocal fold scarring to Rett syndrome. In my doctoral studies, I developed table-top microscopes and endoscopic probes for treatment of scarred vocal folds. First, I determined ablation and imaging parameters for nonlinear imaging guided ultrafast laser surgery with healthy and scarred tissues. Then, I developed a semi-automated system to test feasibility of injecting polyethylene glycol based biomaterial into a laser ablated subsurface void. Finally, I developed a miniaturized ultrafast laser microsurgery probe for vocal fold surgery. In my postdoctoral training, I have developed various two-photon microscopes and a three- photon microscope to perform imaging and modulating of neurons in awake mice as well as imaging intact cerebral organoids. First, I developed a two-photon microscope to perform simultaneous two-color imaging to determine sensory processing through inhibitory interactions between Somatostatin and Parvalbumin interneurons. Second, I developed an optimized three-photon microscope to image evoked neuronal responses through all cortical layers and subplate of visual cortex in awake mice as well as to image intact cerebral organoids for Rett syndrome. In the third project, I developed a wide-field two-photon microscope to image and modulate neurons in the primary and higher visual areas of behaving mice. Overall, my education and research experience establish my long-term research goal to develop next-generation multiphoton systems to reveal both structural and functional connectivity in various biological systems for diagnosing and treating disorders.

Education: • The University of Texas at Austin, August 2015. PhD, Mechanical Engineering • Middle East Technical University, June 2009. MSc, Mechanical Engineering • Middle East Technical University, June 2006. BS, Mechanical Engineering

Research/Work Experience: • Massachusetts Institute of Technology, September 2015-present Postdoctoral Associate, Picower Institute for Learning and Memory, Advisors: Mriganka Sur and Peter TC So • The University of Texas at Austin, January 2010- August 2015 Graduate Research Assistant, Mechanical Engineering, Advisor: Adela Ben-Yakar • Middle East Technical University, September 2006- December 2009 Graduate Research Assistant, Mechanical Engineering, Advisor: Zafer Dursunkaya • Robert Bosch GmbH, Summer 2005. Intern. Research and Development

Selected Publications: 1. Yildirim M, Sugihara H, So PTC, Sur M. Functional imaging of visual cortical layers and subplate in awake mice with optimized three-photon microscopy. Nature Communications (2019); 10(1):177. 2. Rikhye R, Hu M, Yildirim M, Sur M. Reliable sensory processing in mouse visual cortex through inhibitory interactions between Somatostatin and Parvalbumin interneurons. Nature Communications (in press). 3. Yildirim M, Delepine C, Pham V, Feldman D, Chou S, So PTC, Sur M. Three-photon imaging of intact human cerebral organoids to assess key components of early neurogenesis in Rett Syndrome. Nat. Methods, in review. 4. Yildirim M, Durr N, Ben-Yakar A. Tripling the maximum imaging depth with third-harmonic generation microscopy. Journal of Biomedical Optics (2015); 20 (9), 096013-096013. 5. Yildirim M, Ferhanoglu O, Kobler J, Zeitels SM, Ben-Yakar A. Parameters Affecting Ultrafast Laser Microsurgery of Subepithelial Voids for Scar Treatment in Vocal Folds. J Biomed Opt (2013); 18(11), 118001-1180014.

Awards/Honors: • Picower Institute for Learning and Memory Engineering Collaboration Grant, 2018-2020. • Best Poster Presentation Award, International Conference on Optics of Surface and Interfaces, 2015. • Best Poster Presentation Award, NEBEC Conference, Boston University, 2014. • Jen-Lab Young Investigator Award for Best Paper, SPIE Photonics West Conference, 2013. KYOKO YOSHIDA, PhD Biomedical Engineering, University of Virginia, MR5, Room 2232, 415 Lane Road, Charlottesville, Virginia, 22908 [email protected]

Research Overview: Pregnancy stands at the interface of biology and mechanics. Throughout pregnancy, circulating hormone levels surge while the growing fetus continuously loads the maternal organs. In response, maternal soft tissues undergo extensive growth and remodeling (G&R) including a 1000-fold increase in uterine cavity volume. My research focuses on combining experimental and computational approaches to investigate how interactions between biological and mechanical cues drive soft tissue growth, remodeling, and mechanical function in an effort to reduce the incidence of preterm birth and maternal mortality due to cardiovascular complications. My Ph.D. focused on understanding how the cervix softens during pregnancy to prepare for delivery. I proposed, fit, and validated a microstructure-based material model against mechanical tests of nonpregnant and pregnant mouse cervices. Overall, I quantified a 4-order of magnitude decrease in cervical stiffness within 18 days. I also collaborated with clinicians and molecular biologists to demonstrate the ability of the pregnancy hormones progesterone and estrogen to remodel collagen and elastic fibers in the cervix. My work laid the foundation for a hormone-mediated computational framework to elucidate this dramatic cervical remodeling process. Stretch and hormones are known drivers of uterine cell G&R, which alter in vivo stretch/stress, which feedback to alter cell behavior. By using computational models, my goal is to elucidate how these complex interactions allow the uterus to G&R without contracting until the baby reaches full term. Because G&R models are still uncommon in pregnancy and because the heart is another muscular organ that undergoes reversible G&R during pregnancy, I focused on cardiac G&R for my postdoctoral fellowship. I first investigated the role of mechanical unloading in triggering reverse remodeling of the heart, a process that occurs not only after pregnancy but also following repair of stenotic or regurgitant heart valves. I demonstrated that approaches which are successful in predicting cardiac growth following hemodynamic overload do a poor job of predicting reverse remodeling and proposed an alternate approach to capture both growth and reversal. I am building on this work to develop a multiscale model, which incorporates signaling pathways related to and the effects of pregnancy hormones on these pathways to determine the synergistic effects of hormones and mechanics on heart growth during pregnancy. I will apply these same techniques to build a model for uterine growth and contractility, which I will use to test new therapeutic targets for preterm birth in the future.

Education: Ph.D., Mechanical Engineering, 2016, Columbia University M.S., Mechanical Engineering, 2010, Columbia University B.S., Mechanical Engineering, 2008, University of Notre Dame

Research/Work Experience: Postdoctoral Research Associate, 2016 – Present, University of Virginia, Biomedical Engineering, Advisor: Jeff Holmes, M.D., Ph.D. Graduate Research Assistant, 2010 – 2016, Columbia University, Mechanical Engineering, Advisor: Kristin Myers, Ph.D. Graduate Research Assistant, 2009 – 2010, Columbia University, Mechanical Engineering, Advisor: Nabil Simaan, Ph.D. Research Student, 2008 – 2009, Tokyo Denki University, Intelligent Mechanical Engineering, Advisor: Yukio Saito, Ph.D.

Selected Publications: K Yoshida, J Omens, A McCulloch, J Holmes “Computational model of left ventricular growth reversal following release of pressure overload”, Submitted, 2019. K Yoshida, C Jayyosi, N Lee, M Mahendroo, K Myers, “Mechanics of cervical remodeling: Insights from rodent models of pregnancy”, Interface Focus 9(5), 2019. S Nallasamy, K Yoshida, M Akins, K Myers, R Iozzo, M Mahendroo, “Steroid hormones are key modulators of tissue mechanical function via regulation of collagen and elastic fibers”, Endocrinology 158(4):950-62, 2017. K Yoshida, J Vink, R Wapner, M Mahendroo, K Myers, “Material properties of mouse cervical tissue in normal gestation”, Acta Biomat 36:195-209, 2016. K Yoshida, C Reeves, J Vink, J Kitajewski, R Wapner, H Jiang, S Cremers, K Myers. Cervical collagen network remodeling in normal pregnancy and disrupted parturition in Antxr2 deficient mice, J Biomech Eng 136(2):21017, 2014.

Awards/Honors: Weinig Scholarship, Columbia University, 2014 – awarded annually to one engineering graduate student 1st place, Ph.D. Student paper competition, ASME Summer Bioengineering Conference, 2013 National Science Foundation Graduate Research Fellowship, 2010 JENNIFER L. YOUNG, PhD Dept. of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, DE [email protected]

Research Overview: My research in the Department of Cellular Biophysics at the Max Planck Institute of Medical Research focuses on understanding how nanoscale extracellular matrix properties influence cancer cells. To do this, I am combining skills I honed during my doctoral research (biomaterials synthesis, cell-matrix interactions) with techniques I have learned in my postdoctoral lab (nanofabrication, cancer cell biology). During my PhD, I focused on characterizing native tissue properties, developing materials to mimic them, and analyzing cellular responses to these new materials. In my postdoctoral studies, I have been able to ‘zoom in’ to the nanoscale by taking advantage of specialized techniques including block copolymer micelle nanolithography, transfer nanolithography, and cryo-SEM, with the ultimate goal of understanding how nanoscale properties of biomaterials influence cell behavior. Specifically, I have investigated the phenomena of acquired chemoresistance in cancer cells, then built nanostructured materials to study the effects of dimensionality and stiffness on this process, again matching biological observations with informed biomaterial synthesis. Throughout my postdoctoral research, I have also been involved in a number of interdisciplinary, international collaborations spanning stem cell differentiation studies, dynamic ligand surface development, and cellnanowire interactions. These experiences will all contribute to my future goal of studying native tissue organization from a nanoscale and materials perspective, and subsequently using this knowledge to develop biologically-relevant materials for investigating a wide variety of cellular behaviors.

Education: • University of California, San Diego, June 2013. PhD, Bioengineering • University of California, Davis, June 2008. BS, Biomedical Engineering

Research/Work Experience: • Max Planck Institute for Medical Research, 2016 – present Postdoctoral Fellow, Cellular Biophysics; Advisor: Prof. Joachim Spatz • Monash University, Melbourne Center for Nanofabrication, Summer 2018 Visiting Academic, Supported by a DAAD Grant; Advisor: Prof. Nicolas Voelcker • Max Planck Institute for Intelligent Systems, 2014 – 2016 Postdoctoral Fellow, New Materials and Biosystems; Advisor: Prof. Joachim Spatz • University of California, San Diego, 2008 – 2013 Graduate Researcher, Bioengineering; Advisor: Prof. Adam Engler • University of California, Davis, 2007 – 2008 Undergraduate Researcher, Biomedical Engineering; Advisor: Prof. J. Kent Leach

Selected Publications: 1. Di Russo, J., Young, J.L., Balakrishnan, A., Benk, A., and Spatz, J.P. NTA-Co3+-His6 versus NTA-Ni2+-His6 mediated E-Cadherin Surface Immobilization Enhances Cellular Traction. Biomaterials. 192: 171-178 (2019). 2. Hadden, W.J.*, Young, J.L.*, Holle, A.W.*, McFetridge, M.L., Kim, D.Y., Wijesinghe, P., Taylor-Weiner, H., Wen, J.H., Lee, A.R., Bieback, K., Vo, B-N., Sampson, D.D., Kennedy, B.F., Spatz, J.P., Engler. A.J., and Choi, Y.S. Stem cell migration and mechanotransduction on linear stiffness gradient hydrogels. PNAS. 114 (22): 5647-5652 (2017). *equal contribution 3. Young, J.L., Holle, A.W., and Spatz. J.P. Nanoscale and mechanical properties of the physiological cell-ECM microenvironment. Exp Cell Res. 343(1): 3-6 (2015). 4. Young, J.L., Kretchmer, K., Ondeck, M., Zambon, A., Engler, A.J. Mechanosensitive Kinases Regulate Stiffness-Induced Cardiomyocyte Maturation. Scientific Reports. 4 (6425) (2014). 5. Young, J.L. and Engler, A.J. Hydrogels with Time-Dependent Mechanical Properties Enhance Cardiomyocyte Differentiation In Vitro. Biomaterials. 32(4): 1002-1009 (2011).

Awards/Honors: • Max Planck Institute Postdoctoral Fellowship, 2014-2017 • American Heart Association Pre-Doctoral Fellow, 2010-2012 • ARCS Pre-Doctoral Fellowship, 2009-2013 STEVEN ZANGANEH, PhD Senior Scientist Center for Translation of Cancer Nanomedicine, Sloan Kettering Institute for Cancer Research, New York Adjunct Faculty, New York University (NYU) Former NIH T32 Postdoc at the Stanford University School of Medicine, Stanford, CA [email protected] Phone: (530) 304-6690

Research/Teaching Interests: My research and teaching goals are directed toward developing Clinical Translational nanoscale technologies with particular emphasis on developing nanoscale biomaterials and immunoengineering systems for immuno-oncology, immunoengineering, and molecular imaging. My main research interest lies in immuno-oncology of solid tumors, with a focus on myeloid cells, particularly tumor-associated macrophages, dendritic cells, and myeloid- derived suppressor cells. A major question my research is investigating is, “What role do nanoparticles play in the tumor microenvironment to regulate innate and adaptive immune systems?” Thus, I am studying the impact of proteins and signals that promote the activation and migration of pro-inflammatory cells into tumor microenvironment and also affect the phenotype of immune cells. In addition, I study human antigen presenting cells (APCs), chemokines, and myeloid cells in linking innate and adaptive immunity. My focus has been on dynamic interactions of macrophages and dendritic cells with T cells during antigen presentation and the possibility of using nanoparticles in immuno-oncology to prime T cells for immune checkpoint inhibitors and CAR-T Cell Therapy. During my postdoctoral fellowship, I developed a state- of-the-art nano-immunotherapy approach for cancer treatment. Outcomes from this discovery have been published in Nature Nanotechnology (Zanganeh et al, 2016 Nov;11(11):986- 994) and is being developed for translation into the clinics. My commitment to high-quality research and teaching is evidenced by my strong publication record (more than 45 peer-reviewed papers that have been cited over 1500 times) and the diverse teaching experiences I have pursued during my graduate and postdoctoral training period.

Education: Stanford Medical School. Dec 2016, Postdoctoral scholar University of Connecticut. May 2014, PhD. Biomedical Engineering K.N.Toosi University of Technology, 2009, Master of Science. Nanomedicine Azad University, BS, Materials Science (Nanomedicine)

Teaching and Mentoring: Adjunct Faculty, New York University (NYU). 2019-Present Bioengineering Technology Entrepreneurship Instructor, Stanford, Sep 2014-Sep 2016 Hume Grant Writing Coach, Stanford Grant Writing Academy, Stanford School of Medicine, Jul 2015. Teaching Certificate from “Learning to Teach” professional development workshop for graduate students, held in Graduate and Postdoctoral Studies Center at University of Connecticut, Nov 2012. Teaching Assistant (2004-2012)

Selected Publication: Iron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissues. NATURE NANOTECHNOLOGY S. Zanganeh, Gregor Hutter, Ryan Spitler, Stuart Goodman, Michael Moseley, Lisa M. Coussens, Heike E. Daldrup-Link, Nature Nanotechnology. 2016 Nov;11(11):986-994

Grants/Fellowships Pending NCI/R01 grant (34th percentile) entitled “Cell Sex and Disease State: Mechanistic Understanding of the Role of Sex in Cancer Nanotechnology.” NIH T32 Postdoctoral Fellowship, The Molecular Imaging Program at Stanford (MIPS), Stanford University 2014-2016 Crow Innovation Grant, University of Connecticut 2012-2013 (10K) Iran Nanotechnology Initiative Council Grant 2008 (50K) FAN ZHANG, PhD Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA, 98109 . [email protected]

Research Overview: The innate immune system is implicated in an enormous number of processes such as malignancy, inflammation, and tissue regeneration that affect millions of people worldwide. Precise targeting of innate immunity is still a challenge, as therapies can often be a double-edged sword and cause either systemic immune suppression or serious autoimmune/non- specific inflammation. Additionally, biomarkers to assess the innate immunity as a readout of disease status or treatment effect are still lacking on the clinical side. There is great potential to apply nanotechnology-based approaches in these processes. The overarching goal of my research is to establish a fundamental understanding of nanodevice design and the ways the devices interact with the innate immune system, with a focus on cancer and inflammatory diseases. There are two major arms in my research program: (1) use nanoparticles as molecular probes to image and analyze innate immunity; and (2) develop nanodevices that deliver RNA-based gene therapy and small molecule-based immune modulators to precisely modulate the innate immunity. My knowledge of engineering and immunology, and my experience of working closely with physicians have placed me in a unique position to establish an interdisciplinary research program that integrates engineering of nanoparticle platforms within the nexus of nanoparticle formulation, immunology, cancer biology, molecular biology, and translational medicine. During my graduate training, I developed engineering and chemistry skills to formulate a dendrimer-based nanomedicine for treating neurological disorders, which is now a patented technology. During my postdoctoral training, I pioneer a research avenue that focused on developing next-generation immunotherapy by genetically programming macrophage through transcriptional regulation. My postdoctoral training solidified my foundation in immunology, molecular biology, cancer biology, and clinically relevant tumor models, leading to a deeper understanding of immunotherapy and gene therapy. Based on my combined doctoral and postdoctoral work, I have published 19 peer-reviewed publications, 4 international patents, and was awarded the Basic Research Award by American Brain Tumor Association, an award that provides $100K over two years.

Education: Ph.D., 2016, Johns Hopkins University, Materials Science and Engineering B.S., 2010, Donghua University, Polymer Engineering

Research/Work Experience: 09/2016-current Fred Hutchinson Cancer Research Center, Seattle, WA, Post-Doctoral Research Fellow 01/2018-08/2018 University of Washington – Seattle, Science Teaching Experience for Postdocs (STEP) fellow, 12/2011-06/2016 Johns Hopkins University, Baltimore, MD, Graduate Research Assistant

Selected Publications: [1] Zhang F, Parayath N, Coon M, Stephan SB, Ene CI, Holland EC, Stephan MT. Genetic programming of macrophages to perform anti-tumor functions using targeted mRNA nanocarriers. Nature Commun. 10, Article number: 3974 (2019) [2] Zhang F, Stephan SB, Ene CI, Smith TT, Holland EC, Stephan MT. Nanoparticles that reshape the tumor milieu create a therapeutic window for effective T cell therapy in solid malignancies. Cancer Research. 2018. 1;78(13):3718-3730. [3] Zhang F*, Mastorakos P*, Mishra M, Mangraviti A, Hwang L, Zhou J, Hanes J, Brem H, Olivi A, Tyler B, Kannan RM. PAMAM Dendrimer Biodistribution in Glioblastoma and Intrinsic Targeting of Tumor Associated Macrophages. Biomaterials. 2015. 52:507-16. [4] Zhang F, Nance E, Alnasser Y, Kannan RM, Kannan S. Microglia migration and interactions with dendrimer in brain in the presence of neuroinflammation. J Neuroinflammation. 2016 Mar 22;13(1):65. [5] Zhang F, Lin YA, Kannan S, Kannan RM. Targeting Specific Cells in the Brain with Nanomedicines for CNS Therapies. J Control Release. 2016. 240:212-226.

Awards/Honors: [1] ABTA Basic Research Fellowships, $100K, 2 years, American Brain Tumor Association, 2018, ongoing [2] Young Investigator Award, Society for Immunotherapy of Cancer 34th Annual Meeting, 2019 [3] BEST POSTER AWARD, Gordon Research Conference on Cancer Nanotechnology, 2019 [4] Young Investigator Research Award, 5nd Annual Immuno-Oncology Young Investigators' Forum (PhD/Postdoctoral Researcher Category, 3rd place), 2019 [5] AACR Scholar-in-training Award, American Association for Cancer Research, 2019 [6] Fred Hutch Student/Postdoc Advisory Committee Travel Award, 2019 JANE Y. ZHANG Mechanical Engineering, University of Washington, 616 NE Northlake Place, Suite 620, Seattle, WA, 98105 [email protected]

Research Overview: My most recent role as an Acting Assistant Professor in University of Washington allowed me to craft my independent research program around close-loop near-patient diagnostics, especially building low-cost molecular diagnostic tools to support patient selftesting and strengthening treatment support. My most recent publications on whole blood HIV sample preparation (especially instrument-free viral lysis and RNA extraction), and ultrasensitive amplification-based HIV prophylaxis therapy adherence monitoring have been accepted to prestigious conferences including MicroTAS, Adherence, BMES, and Gordon Research Conferences. While building my own research programs, I strive to bring the experience from industry to best design and manage two R01 funded programs in Dr. Jonathan Posner and Dr. Paul Drain’s labs. I believe I’m in a great position to launch my own research programs if given an opportunity with any major R1 institutes.

Education: Ph.D., Biomedical Engineering, 2012, Boston University B.A.Sc., Engineering Science, 2005, University of Toronto

Research/Work Experience: 2018-19 Acting Assistant Professor, University of Washington, Seattle, WA 2016-18 Senior Manager, Amgen, Los Angeles, CA 2014 Senior Scientist and Project Lead, Chromologic, Los Angeles, CA 2011-12 Senior Scientist – Post doctoral, BD, RTP, NC

Selected Publications: 1. J.Y. Zhang, A. Olanrewaju, A. Bender, P. Drain, J. Posner, “An Ultra-Sensitive Nucleic Acid Assay for Rapid Measurement of Adherence to HIV Therapy”, BMES, Philadelphia, PA, October 2019. 2. J.Y. Zhang, A. Olanrewaju, A. T. Bender, Y. Zhang, P. Drain, J. Posner, “Ultrasensitive, Semi-Quantitative Measurement of HIV Nucleoside Reverse Transcriptase Inhibitors (NRTI) content in Pre-exposure Prophylaxis (PrEP) Adherence Monitoring with RTRecombinase Polymerase Amplification (RPA)”, μTAS, Basel, Switzerland, October 2019. 3. J.Y. Zhang, M. Mahalanabis, L. Liu, J. Chang, N. Pollack, and C.M. Klapperich, “A Disposable Microfluidic Virus Concentration Device Based on Evaporation and Interfacial Tension”, Diagnostics, 2013, 3(1), 155-169; DOI:10.3390/diagnostics3010155. 4. J.Y. Zhang, Q.Q. Cao, C.M. Klapperich, “Integrative Microfluidic Sample Preparation for Chip-based Molecular Diagnostics” chapter in book “Microfluidic Applications for Human Health”, U. Demirci, R. Langer, A. Khademhosseini, and J. Blander eds., World Scientific Publishing Co., Hackensack, NJ, 2012. 5. J.Y. Zhang, J. Do, C.M. Klapperich, “Rapid Point-of-Care Concentration of Bacteria in a Disposable Microfluidic Device Using Meniscus Dragging Effect”, Lab Chip, 2010, DOI: 10.1039/c0lc00051e. 6. J.Y. Zhang, A. Ergin, K.L. Andken, C. Sheng, I.J. Bigio, “Design of a Dynamic Optical Tissue Phantom to Model Extravasation Pharmacokinetics”, Proc. SPIE, Vol. 7567, 75670J, 2010, DOI: 10.1117/12.840547 7. J. Do, J.Y. Zhang, and C.M. Klapperich, “Maskless Writing of Microfluidics: Rapid Prototyping of 3D Microfluidics Using Scratch on a Polymer Substrate,” Robotics and Computer Integrated Manufacturing, 2010, DOI: 10.1016/j.rcim.2010.06.004. 8. J.Y. Zhang, Y.K.S. Lee, Y. Zhang, U. S. Patent 62/651,626: “Otoscope Pen”, Provisional filed April 2, 2018. 9. J.Y. Zhang, Y.K.S. Lee, Y. Zhang, U. S. Patent 62/648,866: “Mobile device adaptive otoscope”, Provisional filed March 27, 2018. 10. J.Y. Zhang, Y.K.S. Lee, Y. Zhang, PCT/US2019/025437: “Portable Otoscope”, Filed April 2, 2019.

Awards/Honors: 2019 FDA P50FD006428 (Subaward PI), Pediatric Ear Monitor for Use by Parents 2018 UCLA Muse Innovation Fellowship, Knapp Venture Competition Finalist, Deutschman Venture Fellowship 2017 NIH R43 AI122497 (Co-PI), Rapid Paper-based Diagnostics of CT/Trich 2015 NIH R43 ES025526 (Proposing PI), Rapid Biomonitoring of Volatile Toxins in Human Samples 2015 NIH R43 272201500020C (Proposing PI), Simple, Inexpensive Unit for Removing Cells from Small Amounts of Blood in Resource-Limited Settings 2008-09 CIMIT-BU Medical Engineering Graduate Student Fellowship, CIMIT Innovation Congress 1st Place Poster Award YANG ZHANG, PhD Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208 [email protected]

Research Overview: Understanding of biomolecular dynamics in the context of the highly complex nanoscale environments are opening up the next frontier for biological discovery. However, it remains challenging to image these biomolecular assemblies, their function and interactions in structurally and dynamically complex cellular systems. To fill this technical void, my research will focus on the development of (1) fluorescent probes and (2) imaging methods for non-invasive nanoscale imaging tools integrating with machinelearning to understand (3) the structure and dynamics in life process. My PhD research covers fluorescence study, organic synthesis and supramolecular chemistry. I invented photoswitchable dyes with infinite contrast for multiplexed fluorescence imaging, cancer cell detection, dynamic tracking in vivo Additionally, I developed ultra-bright fluorescent supramolecular nanocarriers for the intracellular delivery of guest cargo. My postdoctoral research extends to the optical implementation, imaging and processing method development for spectroscopic single-molecule localization microscopy (sSMLM). This innovative imaging technique enables parallel, multiplexed super-resolution imaging. Using sSMLM, I investigated biomolecular dynamics in nuclear pore complexes in differentiation process and post-translational modification in programmed cell apoptosis. These research experiences allow me to fully understand the practical need in specific biological questions as well as the limitations of up-to-date dyes for super-resolution microscopy. With these practical guidelines in mind and the multidisciplinary skills on hand, my research program will emphasize on the development of multidimensional nanoscale imaging tools to visualize complex molecular dynamics.

Education: PhD, University of Miami, Coral Gables, FL 2017 BS, Qilu University of Technology, Jinan, Shandong, China 2012

Research/Work Experience: 1.Northwestern University, Evanston, IL Postdoctoral Fellow; Advisor: Prof. Hao F. Zhang 06/2017-present Development of spectroscopic single-molecule localization microscopy (sSMLM) 2. University of Miami, Coral Gables, FL Graduate Research Assistant; Advisor: Prof. Francisco M. Raymo 08/2012-05/2017 Design photochemical strategies to monitor cellular dynamics in living organism

Selected Publications: (Total number of publications = 29, First/co-first authored publications = 11) 1. Y. Zhang, K. Song, S. Tang, L. Ravelo, J. Cusido, C. Sun, H. F. Zhang, and F.M. Raymo, Far-Red Photoactivatable BODIPYs for the Super-Resolution Imaging of Live Cells. J. Am. Chem. Soc., 2018, 140, 12741-12745. 2. Y. Zhang, K. Song, B. Dong, J. Davis, G. Shao, C. Sun, H. F. Zhang, Multi-color Super-Resolution Imaging using spectroscopic single-molecule localization microscopy. Applied Optics, 2019, 58, 2248-2255. 3. S. Tang#, Y. Zhang#, P. Dhakal#, L. Ravelo, C. L. Anderson, K. M. Collins, F. M. Raymo, Photochemical Barcodes. J. Am. Chem. Soc., 2018, 140,4485-4488. (# Equal Author Contribution) 4. Y. Zhang, S. Swaminathan, S. Tang, J. Garcia-Amorós, M. Boulina, B. Captain, J.D. Baker, and F. M. Raymo, Photoactivatable BODIPYs Designed to Monitor the Dynamics of Supramolecular Nanocarrier. J. Am. Chem. Soc., 2015, 137, 4709–4719 5. Y. Zhang, S. Tang, L. Sansalone, J. D. Baker, F. M. Raymo, A Photoswitchable Fluorophore for the Real-Time Monitoring of Dynamic Events in Living Organisms. Chem. Eur. J.,2016. 22, 15027-15034.

Awards/Honors: Postdoctoral Professional Development Award, Northwestern University (2019) Best Poster Award, International Symposium on Photochromism (2016) Sam and Clara Schreiber Research Fellowship, University of Miami (2016) GAFAC Award, University of Miami (2013,2015) First-Class Scholarship, Qilu University of Technology (2011) JOSHUA ZIMMERMANN, PhD Bioengineering, University of California, Berkeley, Stanley Hall, Berkeley, California, 94720 . [email protected] Research Overview: Organoids hold great promise as in vitro model systems that could vastly advance our understanding of human-specific aspects of disease and development. However, the inherent inhomogeneity that arises from reliance on spontaneous morphogenesis hinders the successful translation of these models. Therefore, my long-term research goal is to engineer technologies that elucidate mechanisms and pathways that guide organization and patterning of stem cells within these 3D organoid cultures. In doing so, I aim to improve the homogeneity and reproducibility of these tissue models and ultimately allow for scale-up and application of this technology for drug screening, disease modeling, and tissue engineering applications. My initial interest in developing 3D tissue models stems from my doctoral research with Dr. Todd McDevitt at the Georgia Institute of Technology and the Gladstone Institute. Through my doctoral work, I established that simply transitioning mesenchymal stem cells (MSCs) from two-dimensional, tissue-culture plastic to threedimensional spheroids imparts vast biological changes in these cells. In particular key paracrine factors critical for MSC immunomodulation were found to be upregulated in 3D spheroids. Through RNA-sequencing and transcriptome analysis, I elucidated key pathways regulating MSC paracrine activity in 3D cultures. Finally, by designing biomaterials for controlled delivery of specific cytokines, I was able to temporally control MSC immunomodulation in spheroids to enhance MSC suppression of T-cell activation. These studies highlight the importance of 3D cultures as both a model platform as well as a means of modulating stem cell behavior and has major implications for the clinical application of MSCs for treatment of inflammatory and immune diseases. For my postdoctoral training, my objective was to further advance my expertise in stem cell engineering and genome editing to develop methods for precise control of stem cell differentiation and morphogenesis. In Dr. David Schaffer’s lab, I have been applying cell engineering techniques to spatiotemporally control developmental signaling pathways to elucidate mechanisms of spatial patterning and to better control these processes. I have been working with models of gastrulation as well as a central nervous system organoid platform to understand emergence of patterning and higher order tissue structures and improve the homogeneity and complexity of these 3D tissue models. Finally, in collaboration with Dr. Douglas Clark’s lab, I have worked with high-throughput screening platforms to rapidly investigate the effects of hundreds of independent culture conditions on differentiation of oligodendrocyte precursors from human pluripotent stem cells. Through this collective training, my experiences in stem cell biology, biomaterials, and cell engineering have laid a strong foundation for future independent research investigating the mechanisms that guide emergence of stem cell patterning and morphogenesis in stem cell organoid models.

Education: 2011-2016: Ph.D., Biomedical Engineering, Georgia Institute of Technology & Emory University 2006-2011: B.S./M.S., Biomedical Engineering, Case Western Reserve University

Research/Work Experience: 2017-Present: Postdoctoral Scholar, University of California, Berkeley 2011-2017: Graduate Research Assistant, Georgia Institute of Technology and Gladstone Institute

Selected Publications: Zimmermann, J.A., Schaffer, D.V. Engineering Biomaterials to Control the Neural Differentiation of Stem Cells. Brain Research Bulletin, 2019. Repina, N.A., Bao X., Zimmermann, et al. Optogenetic Control of Wnt Signaling for Modeling Early Embryogenic Patterning with Human Pluripotent Stem Cells. bioRxiv, 2019. Zimmermann, J.A., Hettiaratchi, M.H., McDevitt, T.C. Enhanced Immunosuppression of T-Cells by Sustained Presentation of Bioactive IFN-γ within 3D Mesenchymal Stem Cell Constructs. Stem Cells Translational Medicine 2016. Allen, A.B., Zimmermann, J.A., et al. Environmental Manipulation to Promote Stem Cell Survival In Vivo: Use o Aggregation, Oxygen Carrier, and BMP-2 Co-Delivery Strategies. Journal of Materials Chemistry B, 2016. Zimmermann, J.A., McDevitt, T.C. Pre-Conditioning Mesenchymal Stromal Cell Spheroids for Immunomodulatory Paracrine Factor Secretion. Cytotherapy, 2014.

Awards/Honors: Ruth L. Kirschstein Institutional National Research Service Award (T32) Training Grant Appointee, 2019-present National Science Foundation Graduate Research Fellowship, 2012-2016 Integrative Graduate Education and Research Traineeship (IGERT), Georgia Institute of Technology, 2011-2013 PAMELA CABAHUG ZUCKERMAN, PhD MSK Research Ortho Surgery, NYU Bioengineering Institute, 433 FIRST AVENUE, 9th Floor Bioengineering, NY, NY, 10010 [email protected]

Research Overview: I am an engineer-scientist, employing principles in Biomedical Engineering (BME) to discover biological control mechanisms to solve medical problems. In particular, I have been studying bone, and how bone tissue cells respond to mechanical signals to affect its maintenance and remodeling. In my PhD work at the City College of NY (CCNY), and current research at NYU, I have been pursuing solutions to the challenges of disuse or aging. At CCNY, I studied under renowned scientists and engineers Drs. Schaffler, Cowin and Weinbaum. Under Schaffler, I lead the work to establish that osteocyte apoptosis is the controlling factor in disuse osteoporosis. In my PhD Thesis work, I identified an integrin-based mechanotransduction complex in bone cells using superresolution microscopy. Now at NYU, as a postdoctoral fellow in Dr. Alesha Castillo's Lab of Mechanobiology and Regenerative Medicine, I am investigating the role of the chemokine recruitment factor, CXCL12, in bone homeostasis and mechano-response, and also studying causes of age-associated alterations in load-induced bone formation. Ongoing work in these projects aims to find molecular control and cellular signaling pathways for osteogenesis. I aspire to participate in profound scientific and engineering discoveries that improve the orthopedic quality of life for our aging population, as well as to positively impact the education of our youth.

Education: Phd Biomedical Engineering, 2016, City College of New York, New York, NY. MSc Microbiology, 2006, Membrane and Ultra Structure, Hebrew University of Jerusalem, Jerusalem, Israel. BS Biomedical Engineering, 1996, Boston University, Boston, MA

Research/Work Experience: 2016-Present. NYU MSK Research and Biomedical Engineering: Castillo Lab of Mechanobiology and Regenerative Medicine. Projects: (1) Determine the role of osteocyte-derived CXCL12 in skeletal development, homeostasis and regeneration from mechanical loading and in fracture repair (2) Assess age-associated decline in stemness of load-induced bone formation. 2009-2015. CCNY Schaffler Lab. Thesis: A Beta3 Integrin Based Mechanosome in Bone Tissue Osteocytes: Plasticity to Changes in Mechanical Loading; Other Projects: (1) Established the role of osteocyte apoptosis in disuse osteoporosis (2) Evaluated the relationship between the acute immune system, and bone development and quality in TLR4/CD14 deficiency. 2006-2009. AECOM Imaging Specialist, Neuroscience Imaging Core. Identified & classified neuronal stem cell differentiation for pilot projects to treat lysosomal storage disorders. 2003-2006. HUJI Trachtenberg Lab. Thesis: Spiroplasma: Effects of Non-Helical Perturbation on Mechanisms of Motility. 1995-1996: BU Mentored by Kamenetskii. Thesis: Design of PNA with Enhanced Gene-Targeting Potency.

Selected Publications: 1. Cabahug-Zuckerman P, Liu C, Cai C, Mahaffey I, Norman SC, Cole W, and Castillo AB. Site-specific load-induced expansion of Sca-1+Prrx1+ and Sca-1-Prrx1+ cells in adult mouse long bone is attenuated with age. JBMR Plus 2019 accepted. 2. Cabahug-Zuckerman P, Stout RF Jr, Majeska RJ, Thi MM, Spray DC, Weinbaum S, Schaffler MB. Potential role for a specialized Beta3 integrin-based structure on osteocyte processes in bone mechanosensation. J Orthop Res. 2018 Feb;36(2):642-652. 3. Cabahug-Zuckerman, P., Frikha-Benayed, D., Majeska, R.J., Tuthill, A., Judex, S., Schaffler, M.B. Osteocyte Apoptosis Caused by Hindlimb Unloading is Required to Trigger Osteocyte RANKL Production and Subsequent Resorption of Cortical and Trabecular Bone in Mice Femurs. J Bone Miner Res. 2016 Jul;31(7):1356-65. 4. Liu C, Cabahug-Zuckerman P, Stubbs C, Pendola M, Cai C, Mann KA, Castillo AB. Mechanical loading promotes the expansion of primitive osteoprogenitors and organizes matrix and vascular morphology in long bone defects. J Bone Miner Res. 2019 Jan 15. doi: 10.1002/jbmr.3668. 5. Cohen-Krausz, S., Cabahug, P.C., Trachtenberg, S. The Monomeric, Tetrameric, and Fibrillar Organization of Fib: The Dynamic Building Block of the Bacterial Linear Motor of Spiroplasma melliferum BC3. J. Mol. Bio. Jul 2011; 410 (2): 194-213.

Awards/Honors: 2018. Orthopedic Research Society: New Investigator Recognition Award. 2018. 2nd NYU Biomedical and Biosystems Conference: 1st Place Best Poster. 2017-2019. NYU CTSI TL1 Postdoctoral Fellowship. 2013. American Society of Bone and Mineral Research: Young Investigator Award. 2011-15. CCNY Palefsky Scholarship and Lead Mentor