National Academies Press: OpenBook

Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium (2015)

Chapter: Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey

« Previous: Engineering Heart Valve Treatment Strategies for Tomorrow--W. David Merryman
Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×

Biomaterials for Treating Myocardial Infarctions

JASON A. BURDICK AND SHAUNA M. DORSEY
University of Pennsylvania

Biomaterials are gaining attention in the development of biomedical therapies for treating patients after a myocardial infarction (i.e., heart attack). These materials may serve as mechanical restraints, vehicles for the delivery of therapeutics, or 3-dimensional scaffolds for tissue regeneration. This article focuses on one particular class of materials: injectable hydrogels, natural or synthetic water-swollen polymer networks that are a promising therapy to attenuate ventricular remodeling after myocardial infarction. They act both as acellular bulking agents to mechanically stabilize the myocardium and as delivery vehicles for cells and/or therapeutic molecules. Various materials, cells, and therapeutic molecules have demonstrated positive outcomes in the repair of cardiac tissue after infarction and provide insight for future material development and optimization. Further development of injectable hydrogels for cardiac repair will have considerable clinical impact by improving therapies to prevent progression to heart failure.

OVERVIEW OF HEART DISEASE

Heart failure affects almost 23 million individuals worldwide (Bui et al. 2011), and nearly 70 percent of these cases are due to coronary artery disease, which causes myocardial infarction (MI) (Go et al. 2014). MI occurs after coronary artery occlusion, resulting in depletion of nutrients and oxygen to the cardiac tissue and subsequent cell death (Cleutjens and Creemers 2002). The death of cells (i.e., cardiomyocytes) leads to the recruitment of inflammatory cells to remove the necrotic debris and the activation of bioactive molecules such as matrix metalloproteinases, which in turn cause degradation of the extracellular matrix (ECM) in cardiac tissue, weakening the myocardial wall and making it susceptible to global

Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×

geometric changes, including thinning and dilation (Buckberg 2005; Dobaczewski et al. 2010; Holmes et al. 2005; Nahrendorf 2011; Spinale 2007). Infarct expansion occurs after the initial problems and is a progressive pathologic process that causes abnormal stress distributions in the borderzone regions surrounding the infarct. The process, additional cell death, and increases in borderzone stress are termed left ventricular (LV) remodeling and can lead to altered contractile properties and heart failure (Epstein et al. 2002; Jackson et al. 2003; Pilla et al. 2005).

TREATMENT STRATEGIES

Building on understanding of the biological and mechanical processes after MI, many strategies now utilize biomaterials for patient treatment. Several focus on treatment after significant tissue remodeling; for example, with tissue engineering, replacement cardiac tissue is developed in the laboratory and then implanted to replace damaged tissue. Another promising approach involves treating the tissue during the acute phase to try to attenuate the remodeling response before significant damage.

One option is to limit the initial infarct expansion, which has been identified as associated with the LV remodeling that leads to heart failure. Previous strategies to limit infarct expansion involved surgical reconstruction of the dilated LV and physical restraint of the ventricle or infarct region using polymeric meshed materials to prevent dilation (Batista et al. 1997; Klodell et al. 2008; Starling et al. 2007), but these approaches are highly invasive and require open-chest surgery.

Injectable biomaterials are being developed as a minimally invasive alternative to decrease damage to surrounding tissues. Among numerous potentially injectable biomaterials (e.g., microparticles), injectable hydrogels are particularly promising; they are water-swollen networks of polymer chains that have a high degree of tunability and can be formed through numerous crosslinking mechanisms (Ruel-Gariepy and Leroux 2004). They have been shown to mechanically stabilize the myocardial wall and modulate LV remodeling either alone or through the delivery of therapies such as cells and growth factors (Figure 1) (Nelson et al. 2011; Tous et al. 2011).

Acellular Approaches

Many investigators believe that post-MI regional mechanical changes and stresses in the myocardium should be addressed when designing biomaterial-based approaches for cardiac repair (Gupta et al. 1994; Holmes et al. 2005; Nelson et al. 2011). As described by the Law of Laplace (Equation 1), stress (T) is directly proportional to pressure (P) and the radius of curvature (R) and inversely proportional to the myocardial thickness (h). Therefore, the increase in R and decrease in h that occur after MI leads to an increase in T.

Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×

image

(1)

Injectable biomaterials can limit infarct expansion by bulking the damaged myocardial wall through mechanical stabilization (Tous et al. 2011). Infarcts naturally stiffen over time as wound healing progresses and collagen is deposited; modifying the tissue properties of the infarct region before the body compensates for the remodeling process can limit infarct expansion and post-MI remodeling (Tous et al. 2011). Injectable hydrogels act as bulking agents by increasing the myocardial wall thickness (h) to decrease LV dilation (as measured by R) and in turn decrease wall stress (T). Theoretical finite element models have confirmed this mechanism of treatment by demonstrating that hydrogels decrease both LV dilation and myofiber stresses (Wall et al. 2006).

Injectable hydrogels can be grouped into either natural or synthetic materials. Natural materials offer advantages such as inherent biological properties, including receptor-binding ligands and susceptibility to proteolytic degradation

image

FIGURE 1 Injectable hydrogel approaches for the treatment of MI. Hydrogels (shown in the center column) can be used as (A) acellular bulking agents or a vehicle for (B) delivery of cells, (C) therapeutic molecules, or (D) a combination of cells and molecules.

Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×

image

FIGURE 2 Acellular hydrogels as bulking agents for myocardial infarction repair. Injectable hyaluronic acid hydrogel distribution in cardiac tissue explant as shown by (A) magnetic resonance imaging and (B) ex vivo sectioning. Scale bar = 1 cm.

(Karam et al. 2012; Lutolf and Hubbell 2005). For cardiac applications where the goal is to replace or repair the damaged ECM, natural biomaterials more closely mimic features of the native ECM and can also be therapeutic in their degradation products through the recruitment of cells (Sui et al. 2011). Commonly used natural injectable materials for cardiac repair are fibrin, alginate, collagen, Matrigel, chitosan, hyaluronic acid, keratin, and decellularized matrices (Tous et al. 2011). But natural materials have limited tunability in properties.

Synthetic materials have defined material properties such as molecular weight, gelation, hydrophilic/hydrophobic properties, degradation, and mechanics, without batch-to-batch variations (Lutolf and Hubbell 2005). They can also be modified with cell binding sites or adhesive ligands to encourage cell interaction (Davis et al. 2005). Various synthetic materials have been explored for cardiac repair therapy, including poly(N-isopropylacrylamide) (PNIPAm)- and poly(ethylene glycol) (PEG)-based hydrogels (Tous et al. 2011). An example of an injected hydrogel based on hyaluronic acid is shown in Figure 2.

Cellular Approaches

Myocardial infarction results in the loss of over 1 billion cardiomyocytes in the infarct region, and cell delivery is one strategy used for tissue repair (Beltrami et al. 1994). A variety of cell types have been delivered—fetal or neonatal cardiomyocytes, embryonic stem cells (ESCs), skeletal myoblasts, bone marrow–derived stem cells (BSCs), adipose-derived stem cells, and cardiac stem cells (Menasche 2005; Segers and Lee 2008). Each has advantages and disadvantages for use in therapies. For example, ESCs offer the advantage of differentiating into both

Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×

cardiomyocyte and vascular lineages, but their efficacy is limited because of their immunogenicity, risk of tumor development, and ethical concerns (Zimmermann 2011). BSCs are an autologous option that can be readily isolated and delivered to cardiac tissue, but their fate is not clear (Le Blanc and Pittenger 2005).

Although both animal models (Segers and Lee 2008) and clinical studies (Menasche 2005) have demonstrated some enhancement in cardiac function with cell delivery, these improvements are often insufficient and transient, which is believed to result from unsatisfactory cell retention, survival, and engraftment (D’Alessandro and Michler 2010). For example, less than 10 percent of BSCs delivered have been detected two hours after injection (Hofmann et al. 2005; Hou et al. 2005), and of those that stay at the injury site approximately 90 percent die within the first week because of physical stress, ischemia (due to microvasculature obstruction), inflammation, and release of cytokines and reactive oxygen species (Robey et al. 2008).

Injectable hydrogels have been explored to enhance cell retention and engraftment for cardiac repair by improving cell attachment, migration, and survival upon delivery (Huang et al. 2005). They permit both high encapsulation efficiency (cells are entrapped during gelation) and precise control over the biophysical and biochemical microenvironment surrounding cells after delivery (Bian et al. 2009).

As with acellular hydrogels, both synthetic and natural polymers have been investigated. Natural materials, such as fibrin, alginate, collagen, and Matrigel, are a popular choice for cell delivery because their inherent biological activity initiates cell-biomaterial interactions (Tous et al. 2011). Synthetic hydrogels can also be used to deliver cells for cardiac repair. With their tunability, synthetic materials can be modified to control both adhesion for cell retention and degradation for desired timing of cell release into the tissue environment. As with the acellular hydrogels, the primary synthetic materials used for cell delivery are PNIPAm and PEG (Tous et al. 2011).

INJECTABLE HYDROGELS FOR MOLECULE DELIVERY

In addition to the approaches described above to alter local mechanical stabilization and serve as a cell delivery vehicle, injectable hydrogels can deliver therapeutic molecules to address post-MI LV remodeling.

Tissue repair is a complex process controlled in part by numerous molecules, such as growth factors and cytokines, and the delivery of such molecules can modulate post-MI endogenous biological responses (Segers and Lee 2010). Delivery of therapeutic molecules alone, by either direct myocardial injection or systemic intravenous circulation, has helped restore cardiac function in some animal models, but the short half-life of the molecules and off-target complications limit clinical application (Urbanek et al. 2005).

Because of these limitations, injectable hydrogels have been used as delivery vehicles to localize molecules and tailor release kinetics through changes in

Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×

polymer-molecule interactions, polymer hydrophobicity, and hydrogel degradation (Chen and Mooney 2003; Kretlow et al. 2007). Hydrogels can both sustain local molecule release and prolong molecule bioactivity (Langer and Folkman 1976). For cardiac applications, injectable hydrogels are useful to deliver antiapoptotic molecules (which limit cell death after injury), angiogenic factors to promote vessel formation, or chemoattractants to recruit cells for repair and attenuation of post-MI remodeling (Tous et al. 2011).

LOOKING FORWARD

As discussed here, a range of injectable hydrogels, cell types, and molecules have been delivered with the intent of attenuating LV remodeling after myocardial infarction. Although many hydrogels have shown positive outcomes in animal models, only one (alginate) has progressed to clinical trials.1 Research is needed to elucidate the effects of hydrogel properties, mode of delivery (e.g., direction injection vs. catheter delivery), and timing of delivery (e.g., acute vs. chronic MI) on LV remodeling. Future studies should further investigate the mechanisms by which hydrogels act on the heart, including both biological and mechanical effects, and focus on clinically relevant parameters to optimize repair outcomes.

REFERENCES

Batista RJ, Verde J, Nery P, Bocchino L, Takeshita N, Bhayana JN, Bergsland J, Graham S, Houck JP, Salerno TA. 1997. Partial left ventriculectomy to treat end-stage heart disease. Annals of Thoracic Surgery 64(3):634–638.

Beltrami CA, Finato N, Rocco M, Feruglio GA, Puricelli C, Cigola E, Quaini F, Sonnenblick EH, Olivetti G, Anversa P. 1994. Structural basis of end-stage failure in ischemic cardiomyopathy in humans. Circulation 89(1):151–163.

Bian W, Liau B, Badie N, Bursac N. 2009. Mesoscopic hydrogel molding to control the 3D geometry of bioartificial muscle tissues. Nature Protocols 4(10):1522–1534.

Buckberg GD. 2005. Architecture must document functional evidence to explain the living rhythm. European Journal of Cardio-Thoracic Surgery 27(2):202–209.

Bui AL, Horwich TB, Fonarow GC. 2011. Epidemiology and risk profile of heart failure. Nature Reviews Cardiology 8(1):30–41.

Chen RR, Mooney DJ. 2003. Polymeric growth factor delivery strategies for tissue engineering. Pharmaceutical Research 20(8):1103–1112.

Cleutjens JP, Creemers EE. 2002. Integration of concepts: Cardiac extracellular matrix remodeling after myocardial infarction. Journal of Cardiac Failure 8(6 Suppl):S344–S348.

D’Alessandro DA, Michler RE. 2010. Current and future status of stem cell therapy in heart failure. Current Treatment Options in Cardiovascular Medicine 12(6):614–627.

Davis ME, Hsieh PC, Grodzinsky AJ, Lee RT. 2005. Custom design of the cardiac microenvironment with biomaterials. Circulation Research 97(1):8–15.

_____________

1 Information about the trials is available at http://clinicaltrials.gov/ct2/show/NCT01226563 (“IK-5001 for the Prevention of Remodeling of the Ventricle and Congestive Heart Failure after Acute Myocardial Infarction”) and at http://clinicaltrials.gov/ct2/results?term=Safety+and+Feasibility+of+the+Injectable+BL-1040+Implant&Search=Search.

Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×

Dobaczewski M, Gonzalez-Quesada C, Frangogiannis NG. 2010. The extracellular matrix as a modulator of the inflammatory and reparative response following myocardial infarction. Journal of Molecular and Cellular Cardiology 48(3):504–511.

Epstein FH, Yang Z, Gilson WD, Berr SS, Kramer CM, French BA. 2002. MR tagging early after myocardial infarction in mice demonstrates contractile dysfunction in adjacent and remote regions. Magnetic Resonance in Medicine 48(2):399–403.

Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ, Dai S, Ford ES, Fox CS, Franco S, Fullerton HJ, Gillespie C, Hailpern SM, Heit JA, Howard VJ, Huffman MD, Judd SE, Kissela BM, Kittner SJ, Lackland DT, Lichtman JH, Lisabeth LD, Mackey RH, Magid DJ, Marcus GM, Marelli A, Matchar DB, McGuire DK, Mohler ER 3rd, Moy CS, Mussolino ME, Neumar RW, Nichol G, Pandey DK, Paynter NP, Reeves MJ, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Wong ND, Woo D, Turner MB, American Heart Association Statistics Committee and Stroke Statistics Subcommittee. 2014. Heart disease and stroke statistics–2014 update: A report from the American Heart Association. Circulation 129(3):e28–e292.

Gupta K, Ratcliffe M, Fallert M, Edmunds L, Bogen D. 1994. Changes in passive mechanical stiffness of myocardial tissue with aneurysm formation. Circulation 89(5):2315–2326.

Hofmann M, Wollert KC, Meyer GP, Menke A, Arseniev L, Hertenstein B, Ganser A, Knapp WH, Drexler H. 2005. Monitoring of bone marrow cell homing into the infarcted human myocardium. Circulation 111(17):2198–2202.

Holmes JW, Borg TK, Covell JW. 2005. Structure and mechanics of healing myocardial infarcts. Annual Review of Biomedical Engineering 7:223–253.

Hou D, Youssef EA, Brinton TJ, Zhang P, Rogers P, Price ET, Yeung AC, Johnstone BH, Yock PG, March KL. 2005. Radiolabeled cell distribution after intramyocardial, intracoronary, and interstitial retrograde coronary venous delivery: Implications for current clinical trials. Circulation 112(9 Suppl):I150–I156.

Huang NF, Yu J, Sievers R, Li S, Lee RJ. 2005. Injectable biopolymers enhance angiogenesis after myocardial infarction. Tissue Engineering 11(11–12):1860–1866.

Jackson BM, Gorman JH 3rd, Salgo IS, Moainie SL, Plappert T, St John-Sutton M, Edmunds LH Jr, Gorman RC. 2003. Border zone geometry increases wall stress after myocardial infarction: Contrast echocardiographic assessment. American Journal of Physiology: Heart and Circulatory Physiology 284(2):H475–H479.

Karam JP, Muscari C, Montero-Menei CN. 2012. Combining adult stem cells and polymeric devices for tissue engineering in infarcted myocardium. Biomaterials 33(23):5683–5695.

Klodell CT Jr, Aranda JM Jr, McGiffin DC, Rayburn BK, Sun B, Abraham WT, Pae WE Jr, Boehmer JP, Klein H, Huth C. 2008. Worldwide surgical experience with the Paracor HeartNet cardiac restraint device. Journal of Thoracic and Cardiovascular Surgery 135(1):188–195.

Kretlow JD, Klouda L, Mikos AG. 2007. Injectable matrices and scaffolds for drug delivery in tissue engineering. Advanced Drug Delivery Reviews 59(4–5):263–273.

Langer R, Folkman J. 1976. Polymers for the sustained release of proteins and other macromolecules. Nature 263(5580):797–800.

Le Blanc K, Pittenger M. 2005. Mesenchymal stem cells: Progress toward promise. Cytotherapy 7(1):36–45.

Lutolf M, Hubbell J. 2005. Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering. Nature Biotechnology 23(1):47–55.

Menasche P. 2005. Stem cells for clinical use in cardiovascular medicine: Current limitations and future perspectives. Thrombosis and Haemostasis 94(4):697–701.

Nahrendorf M. 2011. Imaging of infarct healing predicts left ventricular remodeling and evolution of heart failure: Focus on protease activity. Circulation: Cardiovascular Imaging 4(4):351–353.

Nelson DM, Ma Z, Fujimoto KL, Hashizume R, Wagner WR. 2011. Intra-myocardial biomaterial injection therapy in the treatment of heart failure: Materials, outcomes and challenges. Acta Biomaterialia 7(1):1–15.

Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×

Pilla JJ, Blom AS, Gorman JH 3rd, Brockman DJ, Affuso J, Parish LM, Sakamoto H, Jackson BM, Acker MA, Gorman RC. 2005. Early postinfarction ventricular restraint improves borderzone wall thickening dynamics during remodeling. Annals of Thoracic Surgery 80(6):2257–2262.

Robey TE, Saiget MK, Reinecke H, Murry CE. 2008. Systems approaches to preventing transplanted cell death in cardiac repair. Journal of Molecular and Cellular Cardiology 45(4):567–581.

Ruel-Gariepy E, Leroux JC. 2004. In situ-forming hydrogels: Review of temperature-sensitive systems. European Journal of Pharmaceutics and Biopharmaceutics 58(2):409–426.

Segers VF, Lee RT. 2008. Stem-cell therapy for cardiac disease. Nature 451(7181):937–942.

Segers VF, Lee RT. 2010. Protein therapeutics for cardiac regeneration after myocardial infarction. Journal of Cardiovascular Translational Research 3(5):469–477.

Spinale FG. 2007. Myocardial matrix remodeling and the matrix metalloproteinases: Influence on cardiac form and function. Physiological Reviews 87(4):1285–1342.

Starling RC, Jessup M, Oh JK, Sabbah HN, Acker MA, Mann DL, Kubo SH. 2007. Sustained benefits of the CorCap Cardiac Support Device on left ventricular remodeling: Three year follow-up results from the Acorn clinical trial. Annals of Thoracic Surgery 84(4):1236–1242.

Sui R, Liao X, Zhou X, Tan Q. 2011. The current status of engineering myocardial tissue. Stem Cell Reviews 7(1):172–180.

Tous E, Purcell B, Ifkovits JL, Burdick JA. 2011. Injectable acellular hydrogels for cardiac repair. Journal of Cardiovascular Translational Research 4(5):528–542.

Urbanek K, Rota M, Cascapera S, Bearzi C, Nascimbene A, De Angelis A, Hosoda T, Chimenti S, Baker M, Limana F, Nurzynska D, Torella D, Rotatori F, Rastaldo R, Musso E, Quaini F, Leri A, Kajstura J, Anversa P. 2005. Cardiac stem cells possess growth factor-receptor systems that after activation regenerate the infarcted myocardium, improving ventricular function and long-term survival. Circulation Research 97(7):663–673.

Wall ST, Walker JC, Healy KE, Ratcliffe MB, Guccione JM. 2006. Theoretical impact of the injection of material into the myocardium: A finite element model simulation. Circulation 114(24):2627–2635.

Zimmermann WH. 2011. Embryonic and embryonic-like stem cells in heart muscle engineering. Journal of Molecular and Cellular Cardiology 50(2):320–326.

Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×
Page 71
Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×
Page 72
Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×
Page 73
Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×
Page 74
Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×
Page 75
Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×
Page 76
Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×
Page 77
Suggested Citation:"Biomaterials for Treating Myocardial Infarctions--Jason A. Burdick and Shauna M. Dorsey." National Academy of Engineering. 2015. Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium. Washington, DC: The National Academies Press. doi: 10.17226/18985.
×
Page 78
Next: Regulatory Perspectives on Technologies for the Heart--Tina M. Morrison »
Frontiers of Engineering: Reports on Leading-Edge Engineering from the 2014 Symposium Get This Book
×
Buy Paperback | $44.00 Buy Ebook | $35.99
MyNAP members save 10% online.
Login or Register to save!
Download Free PDF

This volume presents papers on the topics covered at the National Academy of Engineering's 2014 US Frontiers of Engineering Symposium. Every year the symposium brings together 100 outstanding young leaders in engineering to share their cutting-edge research and innovations in selected areas. The 2014 symposium was held September 11-13 at the National Academies Beckman Center in Irvine California. The topics covered at the 2014 symposium were: co-robotics, battery materials, technologies for the heart, and shale gas and oil. The intent of this book is to convey the excitement of this unique meeting and to highlight innovative developments in engineering research and technical work.

  1. ×

    Welcome to OpenBook!

    You're looking at OpenBook, NAP.edu's online reading room since 1999. Based on feedback from you, our users, we've made some improvements that make it easier than ever to read thousands of publications on our website.

    Do you want to take a quick tour of the OpenBook's features?

    No Thanks Take a Tour »
  2. ×

    Show this book's table of contents, where you can jump to any chapter by name.

    « Back Next »
  3. ×

    ...or use these buttons to go back to the previous chapter or skip to the next one.

    « Back Next »
  4. ×

    Jump up to the previous page or down to the next one. Also, you can type in a page number and press Enter to go directly to that page in the book.

    « Back Next »
  5. ×

    Switch between the Original Pages, where you can read the report as it appeared in print, and Text Pages for the web version, where you can highlight and search the text.

    « Back Next »
  6. ×

    To search the entire text of this book, type in your search term here and press Enter.

    « Back Next »
  7. ×

    Share a link to this book page on your preferred social network or via email.

    « Back Next »
  8. ×

    View our suggested citation for this chapter.

    « Back Next »
  9. ×

    Ready to take your reading offline? Click here to buy this book in print or download it as a free PDF, if available.

    « Back Next »
Stay Connected!