Abstract

Central MessageRapid advances in stem cell medicine and genome editing technology may result in a paradigm shift in heart transplantation.See Commentary on page XXX. Rapid advances in stem cell medicine and genome editing technology may result in a paradigm shift in heart transplantation. See Commentary on page XXX. Heart transplantation remains the definitive therapy for end-stage heart failure; however, widespread access to transplantation is limited by donor shortages.1Dharmavaram N. Hess T. Jaeger H. Smith J. Hermsen J. Murray D. National trends in heart donor usage rates: are we efficiently transplanting more hearts?.J Am Heart Assoc. 2021; 10: e019655Crossref PubMed Scopus (1) Google Scholar In addition to the increasing burden of heart failure secondary to dietary and lifestyle factors,2Virani S.S. Alonso A. Benjamin E.J. Bittencourt M.S. Callaway C.W. Carson A.P. et al.Heart disease and stroke statistics 2020 update: a report from the American Heart Association.Circulation. 2020; 141: e139-e596Crossref PubMed Scopus (2393) Google Scholar there is a growing population of patients with congenital heart disease, including those with prior univentricular repair.3Schilling C. Dalziel K. Nunn R. Du Plessis K Shi W.Y. Celermajer D et al.The Fontan epidemic: population projections from the Australia and New Zealand Fontan Registry.Int J Cardiol. 2016; 219: 14-19Abstract Full Text Full Text PDF PubMed Scopus (79) Google Scholar As such, alternative strategies for treatment of end-stage heart failure are required. Recent breakthroughs in stem cell medicine and genome editing have led to unprecedented research activity in the field of myocardial regeneration.4Takahashi K. Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors.Cell. 2006; 126: 663-676Abstract Full Text Full Text PDF PubMed Scopus (17471) Google Scholar,5Jinek M. Chylinski K. Fonfara I. Hauer M. Doudna J.A. Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity.Science. 2012; 337: 816-821Crossref PubMed Scopus (7791) Google Scholar Generating specialized therapeutic cells, tissue grafts, and even entire organs from stem cells appears to be within our grasp. As such, we are at the cutting edge of what previously seemed impossible, and we will be pushing into unchartered territory as we seek pioneering innovations. The field of stem cell medicine has been revolutionized by the discovery of induced pluripotent stem cells (iPSCs). In 1962, John B. Gurdon6Gurdon J.B. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles.J Embryol Exp Morphol. 1962; 10: 622-640PubMed Google Scholar cloned a functional tadpole from the nucleus of a specialized intestinal cell of a frog, demonstrating that the nucleus of a mature cell could be returned to a pluripotent state. In 2006, Yamanaka and Takahashi4Takahashi K. Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors.Cell. 2006; 126: 663-676Abstract Full Text Full Text PDF PubMed Scopus (17471) Google Scholar discovered 4 transcription factors that can reprogram mature cells into pluripotent stem cells. The resulting iPSCs can be generated from human cells and are capable of differentiating into cell types of all 3 germ layers.7Takahashi K. Tanabe K. Ohnuki M. Narita M. Ichisaka T. Tomoda K et al.Induction of pluripotent stem cells from adult human fibroblasts by defined factors.Cell. 2007; 131: 861-872Abstract Full Text Full Text PDF PubMed Scopus (13869) Google Scholar,8Yu J. Vodyanik M.A. Smuga-Otto K. Antosiewicz-Bourget J. Frane J.L. Tian S. et al.Induced pluripotent stem cell lines derived from human somatic cells.Science. 2007; 318: 1917-1920Crossref PubMed Scopus (7760) Google Scholar In 2012, Gurdon and Yamanaka were jointly awarded The Nobel Prize in Physiology or Medicine. Despite their enormous potential in the field of regenerative medicine, numerous obstacles need to be addressed before iPSCs can be widely utilized in clinical practice. In particular, the potential for tumorgenicity, immunogenicity, and the integration of transplanted cells are of concern.9Yamanaka S. Pluripotent stem cell-based cell therapy-promise and challenges.Cell Stem Cell. 2020; 27: 523-531Abstract Full Text Full Text PDF PubMed Scopus (112) Google Scholar Nonetheless, Yamanaka's discovery laid the foundation from which a new frontier in transplantation can be explored. In combination with the progress in stem cell research, recent advances in the field of genome editing may have significant implications for transplantation. Namely, the discovery of clustered regularly interspaced short palindromic repeats (CRISPR), and CRISPR-associated 9 (Cas9) protein, has transformed the study of molecular life sciences. Derived from ancient bacterial immune pathways, CRISPR-Cas9 technology functions as a form of a molecular machinery that allows editing of virtually any genome sequence.5Jinek M. Chylinski K. Fonfara I. Hauer M. Doudna J.A. Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity.Science. 2012; 337: 816-821Crossref PubMed Scopus (7791) Google Scholar For their work in discovering CRISPR-Cas9 as a tool for genome editing, Jennifer A. Doudna and Emmanuelle Charpentier were jointly awarded the 2020 Nobel Prize in Chemistry. Although CRISPR-Cas9 is a hugely promising technology, several challenges still face its widespread clinical applications. Most importantly, unintended on-target genome damage, as well as off-target effects, risk introducing pathogenic mutations into edited cells.10Kosicki M. Tomberg K. Bradley A. Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements.Nat Biotechnol. 2018; 36: 765-771Crossref PubMed Scopus (45) Google Scholar,11Saha K. Sontheimer E.J. Brooks P.J. Dwinell M.R. Gersbach C.A. Liu D.R. et al.The NIH somatic cell genome editing program.Nature. 2021; 592: 195-204Crossref PubMed Scopus (14) Google Scholar Such mutations could become neoplastic with time, particularly in iPSCs, given their long replicative lifespan. As such, comprehensive methods for screening and detecting genome damage are mandatory before patient transplantation of edited cells and their derived products.10Kosicki M. Tomberg K. Bradley A. Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements.Nat Biotechnol. 2018; 36: 765-771Crossref PubMed Scopus (45) Google Scholar,11Saha K. Sontheimer E.J. Brooks P.J. Dwinell M.R. Gersbach C.A. Liu D.R. et al.The NIH somatic cell genome editing program.Nature. 2021; 592: 195-204Crossref PubMed Scopus (14) Google Scholar Immune-mediated rejection is a critical obstacle in the widespread use of iPSCs and the potential grafts that may be derived from them.9Yamanaka S. Pluripotent stem cell-based cell therapy-promise and challenges.Cell Stem Cell. 2020; 27: 523-531Abstract Full Text Full Text PDF PubMed Scopus (112) Google Scholar A source of donor iPSCs with no (autologous) or reduced (universal donor-derived or genome-edited) immunogenicity is the crucial first step for bioengineered organs in vitro and in vivo. iPSCs created from a patient's own cells provide the incredible possibility of autologous graft and organ transplantation. Despite initial controversies, it is now established that autologous iPSCs do provide the opportunity to develop rejection-free cell therapies.12Deuse T. Hu X. Agbor-Enoh S. Koch M. Spitzer M. Gravina A. et al.De novo mutations in mitochondrial DNA of iPSCs produce immunogenic neoepitopes in mice and humans.Nat Biotechnol. 2019; 37: 1137-1144Crossref PubMed Scopus (35) Google Scholar, 13Zhao T. Zhang Z.-N. Rong Z. Xu Y. Immunogenicity of induced pluripotent stem cells.Nature. 2011; 474: 212-215Crossref PubMed Scopus (1066) Google Scholar, 14Araki R. Uda M. Hoki Y. Sunayama M. Nakamura M. Ando S. et al.Negligible immunogenicity of terminally differentiated cells derived from induced pluripotent or embryonic stem cells.Nature. 2013; 494: 100-104Crossref PubMed Scopus (360) Google Scholar, 15Mandai M. Watanabe A. Kurimoto Y. Hirami Y. Morinaga C. Daimon T. et al.Autologous induced stem-cell–derived retinal cells for macular degeneration.N Engl J Med. 2017; 376: 1038-1046Crossref PubMed Scopus (665) Google Scholar However, the cost per individual patient and the time required to generate sufficient cell quantities or develop grafts for transplantation is currently seen as a prohibitive factor in the utilization of autologous iPSCs.16Chakradhar S. An eye to the future: researchers debate best path for stem cell–derived therapies.Nat Med. 2016; 22: 116-119Crossref PubMed Scopus (30) Google Scholar,17Madrid M. Sumen C. Aivio S. Saklayen N. Autologous induced pluripotent stem cell–based cell therapies: promise, progress, and challenges.Curr Protoc. 2021; 1: e88Crossref PubMed Scopus (3) Google Scholar This is particularly problematic for time-sensitive clinical indications, such as ischemic cardiomyopathy. However, in conditions where the need for transplantation may be foreshadowed several years in advance, such as hypoplastic left heart syndrome, autologous iPSCs offer the prospect of tissue grafts or entire heart transplantation, without the need for lifelong immunosuppression. In hematopoietic stem cell and solid organ transplantation, immune-mediated rejection is mitigated by matching of donor and recipient human leukocyte antigen (HLA) haplotypes. However, given the variety of HLA haplotypes, the logistics required to prepare the necessary quantity of iPSC lines is currently not practical. Alternatively, it has been proposed that iPSC lines derived from donors who are homozygote for common HLA types could cover a substantial proportion of the population. Such an approach would still require significant logistical hurdles to be overcome and would likely be dependent on large-scale collaboration between existing biobanks. For instance, it has been estimated that to establish a biobank of iPSCs that would cover 90% of the Japanese population, 140 unique HLA-homozygous donors would be needed, which would require typing of ∼160,000 individuals.18Okita K. Matsumura Y. Sato Y. Okada A. Morizane A. Okamoto S. et al.A more efficient method to generate integration-free human iPS cells.Nat Methods. 2011; 8: 409-412Crossref PubMed Scopus (1227) Google Scholar For societies with a broader range of ethnicities, this number would increase.19Gourraud P.A. Gilson L. Girard M. Peschanski M. The role of human leukocyte antigen matching in the development of multiethnic "haplobank" of induced pluripotent stem cell lines.Stem Cells. 2012; 30: 180-186Crossref PubMed Scopus (100) Google Scholar,20Taylor Craig J. Peacock S. Chaudhry Afzal N. Bradley J.A. Bolton Eleanor M. Generating an iPSC Bank for HLA-matched tissue transplantation based on known donor and recipient HLA types.Cell Stem Cell. 2012; 11: 147-152Abstract Full Text Full Text PDF PubMed Scopus (248) Google Scholar To supplement biobanking of HLA-homozygous donors, a recent study utilized genome-editing technology to develop pseudohomozygous iPSCs by HLA allele-specific editing of HLA heterozygote donors.21Xu H. Wang B. Ono M. Kagita A. Fujii K. Sasakawa N. et al.Targeted Disruption of HLA genes via CRISPR-Cas9 generates iPSCs with enhanced immune compatibility.Cell Stem Cell. 2019; 24: 566-578.e7Abstract Full Text Full Text PDF PubMed Scopus (153) Google Scholar Although a previous primate study demonstrated a reduced immune response to HLA-matched allogenic iPSC-cardiomyocytes, immunosuppression is still likely to be required, albeit for a shorter duration and at a lower dose.22Kawamura T. Miyagawa S. Fukushima S. Maeda A. Kashiyama N Kawamura A. et al.Cardiomyocytes derived from MHC-homozygous induced pluripotent stem cells exhibit reduced allogeneic immunogenicity in MHC-matched non-human primates.Stem Cell Rep. 2016; 6: 312-320Abstract Full Text Full Text PDF PubMed Scopus (65) Google Scholar This is in part due to the wide variety of minor histocompatibility antigens that are expressed in the cleft of major histocompatibility complex (MHC) antigens, and differ between donor and recipient, even in the case of HLA matching.23Trounson A. Boyd N.R. Boyd R.L. Toward a universal solution: editing compatibility into pluripotent stem cells.Cell Stem Cell. 2019; 24: 508-510Abstract Full Text Full Text PDF PubMed Scopus (7) Google Scholar Furthermore, genome-editing technology can be used to delete the classical class Ⅰ HLA (HLA-A, HLA-B, and HLA-C), either by direct exon targeting or by interfering with the β2-microglobulin gene (β2-microglobulin forms a heterodimer with class Ⅰ HLA and is necessary for class-Ⅰ HLA presentation on the cell surface) (Figure 1, A). With deletion of classical class Ⅰ HLA, the iPSC may avoid cytotoxic T-cells. However, with complete absence of class Ⅰ HLA the iPSC will be targeted by natural killer (NK) cells, through the missing self-response. Moreover, deletion of all classical class-Ⅰ HLA renders a cell incapable of antigen presentation and risks uncontrolled proliferation of cells that are tumorogenic or infected by pathogens. Rather, targeted deletion of HLA-A and HLA-B (Figure 1, B), with retention of a single HLA-C allele, can create iPSCs that reduce both NK cell and cytotoxic T-cell destruction, because HLA-C presentation can also inhibit NK cell activity.21Xu H. Wang B. Ono M. Kagita A. Fujii K. Sasakawa N. et al.Targeted Disruption of HLA genes via CRISPR-Cas9 generates iPSCs with enhanced immune compatibility.Cell Stem Cell. 2019; 24: 566-578.e7Abstract Full Text Full Text PDF PubMed Scopus (153) Google Scholar Furthermore, HLA-C retention provides some capacity for antigen presentation, reducing the risks related to tumorigenesis and infection.21Xu H. Wang B. Ono M. Kagita A. Fujii K. Sasakawa N. et al.Targeted Disruption of HLA genes via CRISPR-Cas9 generates iPSCs with enhanced immune compatibility.Cell Stem Cell. 2019; 24: 566-578.e7Abstract Full Text Full Text PDF PubMed Scopus (153) Google Scholar Of note, given the relative lack of diversity in the HLA-C haplotype, it is estimated that only 12 donors with various HLA-C haplotypes would be required to provide cells that could create iPSCs for most of the world's population.21Xu H. Wang B. Ono M. Kagita A. Fujii K. Sasakawa N. et al.Targeted Disruption of HLA genes via CRISPR-Cas9 generates iPSCs with enhanced immune compatibility.Cell Stem Cell. 2019; 24: 566-578.e7Abstract Full Text Full Text PDF PubMed Scopus (153) Google Scholar Moreover, to overcome NK cell destruction, genome editing can overexpress a single-chain HLA-E/β2-microglobulin fusion, in which the nonclassical HLA-E can inhibit NK cell activity to a degree.24Gornalusse G.G. Hirata R.K. Funk S.E. Riolobos L. Lopes V.S. Manske G. et al.HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells.Nat Biotechnol. 2017; 35: 765-772Crossref PubMed Scopus (218) Google Scholar In addition, the class-II MHC transactivator gene can be disrupted, depleting class-II MHC and, thus, avoiding HLA-DR-activated helper T-cell toxicity.21Xu H. Wang B. Ono M. Kagita A. Fujii K. Sasakawa N. et al.Targeted Disruption of HLA genes via CRISPR-Cas9 generates iPSCs with enhanced immune compatibility.Cell Stem Cell. 2019; 24: 566-578.e7Abstract Full Text Full Text PDF PubMed Scopus (153) Google Scholar Immune cloaking via genome editing, to induce expression of immune checkpoints molecules, is another strategy to inhibit immune-mediated rejection of allogenic iPSCs.25Rong Z. Wang M. Hu Z. Stradner M Zhu M Kong H et al.An effective approach to prevent immune rejection of human ESC-derived allografts.Cell Stem Cell. 2014; 14: 121-130Abstract Full Text Full Text PDF PubMed Scopus (145) Google Scholar The potential clinical applications of iPSCs ranges from cellular therapy to the production of entire organs for transplantation. To date, preclinical and clinical studies have predominantly focused on cellular therapies; however, there has been substantial progress in the pursuit of bioengineered cardiac tissues and organs suitable for transplantation. Previous animal studies, using bone marrow-derived cells injected into myocardial peri-infarct areas, demonstrated an improvement in cardiac function and regeneration of cardiomyocytes.26Orlic D. Kajstura J. Chimenti S. Jakoniuk I. Anderson S.M. Li B. et al.Bone marrow cells regenerate infarcted myocardium.Nature. 2001; 410: 701-705Crossref PubMed Scopus (4612) Google Scholar Although these results generated enormous excitement in the field of cardiac regenerative medicine, they were not always reproducible.27Balsam L.B. Wagers A.J. Christensen J.L. Kofidis T. Weissman I.L. Robbins R.C. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium.Nature. 2004; 428: 668-673Crossref PubMed Scopus (1509) Google Scholar Subsequent studies have raised doubt about both the efficacy and mechanism of adult stem cell therapies for cardiac disease.28Epstein J.A. A time to press reset and regenerate cardiac stem cell biology.JAMA Cardiol. 2019; 4: 95-96Crossref PubMed Scopus (22) Google Scholar, 29Sano S. Sano T. Ishigami S. Ito T. Cardiac stem cell therapy: does a newborn infant's heart have infinite potential for stem cell therapy?.J Thorac Cardiovasc Surg. 2022; 163: 242-247Abstract Full Text Full Text PDF PubMed Scopus (3) Google Scholar, 30Gyöngyösi M. Haller P.M. Blake D.J. Rendon E.M. Meta-analysis of cell therapy studies in heart failure and acute myocardial infarction.Circ Res. 2018; 123: 301-308Crossref PubMed Scopus (50) Google Scholar In a recent study, intramyocardial injection of live or dead stem cells, or a chemical inducer of innate immunity, all resulted in activation of an innate immune response and improvement in cardiac function.31Vagnozzi R.J. Maillet M. Sargent M.A. Khalil H. Johansen A.Z. Schwanekamp J.A. et al.An acute immune response underlies the benefit of cardiac stem cell therapy.Nature. 2020; 577: 405-409Crossref PubMed Scopus (194) Google Scholar As such, the beneficial effects of adult stem cell therapies may be due to activation of the innate immune response, rather than stem cell engraftment and differentiation into cardiomyocytes. Despite the controversies surrounding adult stem cell therapies,28Epstein J.A. A time to press reset and regenerate cardiac stem cell biology.JAMA Cardiol. 2019; 4: 95-96Crossref PubMed Scopus (22) Google Scholar there are ongoing preclinical and clinical studies investigating the potential benefits of iPSC-derived cardiomyocytes as a form of injectable cellular therapy.32Guan X. Xu W. Zhang H. Wang Q. Yu J. Zhang R. et al.Transplantation of human induced pluripotent stem cell-derived cardiomyocytes improves myocardial function and reverses ventricular remodeling in infarcted rat hearts.Stem Cell Res Ther. 2020; 11: 73Crossref PubMed Scopus (17) Google Scholar, 33Peinkofer G. Maass M. Pfannkuche K. Sachinidis A. Baldus S. Hescheler J. et al.Persistence of intramyocardially transplanted murine induced pluripotent stem cell-derived cardiomyocytes from different developmental stages.Stem Cell Res Ther. 2021; 12: 46Crossref PubMed Scopus (1) Google Scholar, 34Shiba Y. Gomibuchi T. Seto T. Wada Y. Ichimura H. Tanaka Y. et al.Allogeneic transplantation of iPS cell-derived cardiomyocytes regenerates primate hearts.Nature. 2016; 538: 388-391Crossref PubMed Scopus (376) Google Scholar Industrial-scale production of human cardiomyocytes from pluripotent stem cells is now possible,35Denning C. Borgdorff V. Crutchley J. Firth K.A. George V. Karla S. et al.Cardiomyocytes from human pluripotent stem cells: from laboratory curiosity to industrial biomedical platform.Biochim Biophys Acta. 2016; 1863: 1728-1748Crossref PubMed Scopus (168) Google Scholar enabling transplantation of clinically relevant numbers of cardiomyocytes (up to 1 billion) following myocardial infarction in a nonhuman primate model.36Chong J.J. Yang X. Don C.W. Minami E. Liu Y. Weyers J.J. et al.Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts.Nature. 2014; 510: 273-277Crossref PubMed Scopus (853) Google Scholar A number of clinical trials involving direct injection of PSC-derived cardiomyocytes in adults with end-stage heart failure are now underway although some safety concerns, particularly arrhythmias, have been documented in preclinical animal studies.36Chong J.J. Yang X. Don C.W. Minami E. Liu Y. Weyers J.J. et al.Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts.Nature. 2014; 510: 273-277Crossref PubMed Scopus (853) Google Scholar,37Romagnuolo R. Masoudpour H. Porta-Sánchez A. Qiang B. Barry J. Laskary A. et al.Human embryonic stem cell-derived cardiomyocytes regenerate the infarcted pig heart but induce ventricular tachyarrhythmias.Stem Cell Rep. 2019; 12: 967-981Abstract Full Text Full Text PDF PubMed Scopus (90) Google Scholar Human iPSCs can be differentiated into the 4 major cell types of the heart (ie, cardiomyocytes, endothelial cells, cardiac fibroblasts, and smooth muscle cells) and used for cardiac tissue engineering in vitro.38Ronaldson-Bouchard K. Ma S.P. Yeager K. Chen T. Song L. Sirabella D. et al.Advanced maturation of human cardiac tissue grown from pluripotent stem cells.Nature. 2018; 556: 239-243Crossref PubMed Scopus (490) Google Scholar, 39Lowenthal J. Gerecht S. If you build it, they will come.Circ Res. 2020; 127: 225-228Crossref PubMed Scopus (1) Google Scholar, 40Yadid M. Oved H. Silberman E. Dvir T. Bioengineering approaches to treat the failing heart: from cell biology to 3D printing.Nat Rev Cardiol. 2022; 19: 83-99Crossref PubMed Scopus (1) Google Scholar Although current protocols for differentiating iPSCs can reliably produce high-yield and high-purity cardiomyocytes,41Palpant N.J. Pabon L. Friedman C.E. Roberts M. Hadland B. Zaunbrecher R et al.Generating high-purity cardiac and endothelial derivatives from patterned mesoderm using human pluripotent stem cells.Nat Protoc. 2017; 12: 15-31Crossref PubMed Scopus (88) Google Scholar,42Lian X. Zhang J. Azarin S.M. Zhu K. Hazeltine L.B. Bao X. et al.Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/β-catenin signaling under fully defined conditions.Nat Protoc. 2013; 8: 162-175Crossref PubMed Scopus (823) Google Scholar achieving sufficient maturation of these cells to facilitate optimal electrical and mechanical continuum remains difficult. Nonetheless, various strategies, including long-term culture,43Lundy S.D. Zhu W.Z. Regnier M. Laflamme M.A. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells.Stem Cells Dev. 2013; 22: 1991-2002Crossref PubMed Scopus (436) Google Scholar co-culture with supportive cell types,44Tiburcy M. Hudson J.E. Balfanz P. Schlick S. Meyer T. Chang Liao M.L. et al.Defined engineered human myocardium with advanced maturation for applications in heart failure modeling and repair.Circulation. 2017; 135: 1832-1847Crossref PubMed Scopus (263) Google Scholar and electrical38Ronaldson-Bouchard K. Ma S.P. Yeager K. Chen T. Song L. Sirabella D. et al.Advanced maturation of human cardiac tissue grown from pluripotent stem cells.Nature. 2018; 556: 239-243Crossref PubMed Scopus (490) Google Scholar and mechanical stimulation45Tulloch N.L. Muskheli V. Razumova M.V. Korte F.S. Regnier M. Hauch K.D. et al.Growth of engineered human myocardium with mechanical loading and vascular coculture.Circ Res. 2011; 109: 47-59Crossref PubMed Scopus (472) Google Scholar have led to improved maturation of cardiomyocytes. In addition to cellular maturity, replicating the anisotropy between and within native cardiomyocytes is necessary to optimize function and prevent maladaptive processes in bioengineered cardiac tissue.40Yadid M. Oved H. Silberman E. Dvir T. Bioengineering approaches to treat the failing heart: from cell biology to 3D printing.Nat Rev Cardiol. 2022; 19: 83-99Crossref PubMed Scopus (1) Google Scholar To achieve this, various bioengineering methods and materials have been utilized, ranging from cyclic stretch46Riegler J. Tiburcy M. Ebert A. Tzatzalos E. Raaz U. Abilez O.J. et al.Human engineered heart muscles engraft and survive long term in a rodent myocardial infarction model.Circ Res. 2015; 117: 720-730Crossref PubMed Scopus (130) Google Scholar to fabricated replicates of the native extracellular matrix (with both natural and synthetic polymers),47Fleischer S. Shapira A. Feiner R. Dvir T. Modular assembly of thick multifunctional cardiac patches.Proc Natl Acad Sci U S A. 2017; 114: 1898-1903Crossref PubMed Scopus (80) Google Scholar,48Capulli A.K. MacQueen L.A. Sheehy S.P. Parker K.K. Fibrous scaffolds for building hearts and heart parts.Adv Drug Deliv Rev. 2016; 96: 83-102Crossref PubMed Scopus (74) Google Scholar hydrogels,49Gao L. Kupfer M.E. Jung J.P. Yang L. Zhang P. Da Sie Y. et al.Myocardial tissue engineering with cells derived from human-induced pluripotent stem cells and a native-like, high-resolution, 3-dimensionally printed scaffold.Circ Res. 2017; 120: 1318-1325Crossref PubMed Scopus (169) Google Scholar and more advanced techniques such as melt electrospinning writing.50Castilho M. Feyen D. Flandes-Iparraguirre M. Hochleitner G. Groll J. Doevendans P.A.F. et al.Electrospinning writing of poly-hydroxymethylglycolide-co-ε-caprolactone-based scaffolds for cardiac tissue engineering.Adv Healthc Mater. 2017; 6: 10Crossref Scopus (82) Google Scholar In animal studies, bioengineered cardiac tissue created from iPSCs have demonstrated evidence of engraftment, vascularization, and electrical coupling to the recipient cardiac tissue, as well as an improvement in cardiac function.51Weinberger F. Breckwoldt K. Pecha S. Kelly A. Geertz B. Starbatty J. et al.Cardiac repair in Guinea pigs with human engineered heart tissue from induced pluripotent stem cells.Sci Transl Med. 2016; 8: 363ra148Crossref PubMed Scopus (132) Google Scholar,52Gao L. Gregorich Z.R. Zhu W. Mattapally S. Oduk Y. Lou X. et al.Large cardiac muscle patches engineered from human induced-pluripotent stem cell-derived cardiac cells improve recovery from myocardial infarction in swine.Circulation. 2018; 137: 1712-1730Crossref PubMed Scopus (178) Google Scholar Furthermore, successful epicardial transplantation of human scale (up to 5 cm × 7 cm) patches in porcine hearts has demonstrated safety, feasibility, and efficacy of such an approach.52Gao L. Gregorich Z.R. Zhu W. Mattapally S. Oduk Y. Lou X. et al.Large cardiac muscle patches engineered from human induced-pluripotent stem cell-derived cardiac cells improve recovery from myocardial infarction in swine.Circulation. 2018; 137: 1712-1730Crossref PubMed Scopus (178) Google Scholar,53Querdel E. Reinsch M. Castro L. Kose D. Bahr A. Reich S. et al.Human engineered heart tissue patches remuscularize the injured heart in a dose-dependent manner.Circulation. 2021; 143: 1991-2006Crossref PubMed Scopus (11) Google Scholar Another study demonstrated increased retention and engraftment of intramyocardially injected cardiomyocytes, when delivered concomitantly with epicardial transplantation of a human mesenchymal stem cell-loaded patch, which provided a complimentary microenvironment and enhanced vascular regeneration.54Park S.-J. Kim R.Y. Park B.-W. Lee S. Choi S.W. Park J. et al.Dual stem cell therapy synergistically improves cardiac function and vascular regeneration following myocardial infarction.Nat Commun. 2019; 10: 3123Crossref PubMed Scopus (77) Google Scholar Despite the progress in bioengineering cardiac tissue, ongoing challenges related to integration, compatibility, sustained viability, and electromechanical coupling with native myocardium remain before successful clinical translation.40Yadid M. Oved H. Silberman E. Dvir T. Bioengineering approaches to treat the failing heart: from cell biology to 3D printing.Nat Rev Cardiol. 2022; 19: 83-99Crossref PubMed Scopus (1) Google Scholar Progress in bioengineering cardiac tissue has meant the prospect of a bioengineered cardiac pump suitable for transplantation is becoming increasingly realistic. However, generating cardiac tissue that is thick enough to withstand the loading forces of the heart and produce sufficient contractile power to supplement or substitute ventricular function remains difficult. In particular, generating a physiological cell density of cardiomyocytes (∼108 cells/cm3) while replicating the geometry of cardiac structures and integrating a vascular network (Figure 2) to ensure viability of thick bioengineered cardiac tissue is challenging.40Yadid M. Oved H. Silberman E. Dvir T. Bioengineering approaches to treat the failing heart: from cell biology to 3D printing.Nat Rev Cardiol. 2022; 19: 83-99Crossref PubMed Scopus (1) Google Scholar,55MacQueen L.A. Sheehy S.P. Chantre C.O. Zimmerman J.F. Pasqualini F.S. Liu X. et al.A tissue-engineered scale model of the heart ventricle.Nat Biomed Eng. 2018; 2: 930-941Crossref PubMed Scopus (82) Google Scholar, 56Taylor D.A. Hochman-Mendez C. Sampaio L.C. Are we close to bioengineering a human-sized, functional heart?.J Thorac Cardiovasc Surg. 2020; 159: 1357-1360Abstract Full Text Full Text PDF PubMed Scopus (2) Google Scholar, 57Fang Y. Sun W. Zhang T. Xiong Z. Recent advances on bioengineering approaches for fabrication of functional engineered cardiac pumps: a review.Biomaterials. 2022; 280: 121298Crossref PubMed Scopus (1) Google Scholar Several approaches exist for tissue vascularization, including natural ingrowth of blood vessels from the host58Sekine H. Shimizu T. Hobo K. Sekiya S. Yang J. Yamato M. et al.Endothelial cell coculture within tissue-engineered cardiomyocyte sheets

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call