Intramyocardial cell-based therapy with Lomecel-B during bidirectional cavopulmonary anastomosis for hypoplastic left heart syndrome: the ELPIS phase I trial

Sunjay Kaushal, Joshua M Hare, Jessica R Hoffman, Riley M Boyd, Kevin N Ramdas, Nicholas Pietris, Shelby Kutty, James S Tweddell, S Adil Husain, Shaji C Menon, Linda M Lambert, David A Danford, Seth J Kligerman, Narutoshi Hibino, Laxminarayana Korutla, Prashanth Vallabhajosyula, Michael J Campbell, Aisha Khan, Eric Naioti, Keyvan Yousefi, Danial Mehranfard, Lisa McClain-Moss, Anthony A Oliva, Michael E Davis, Sunjay Kaushal, Joshua M Hare, Jessica R Hoffman, Riley M Boyd, Kevin N Ramdas, Nicholas Pietris, Shelby Kutty, James S Tweddell, S Adil Husain, Shaji C Menon, Linda M Lambert, David A Danford, Seth J Kligerman, Narutoshi Hibino, Laxminarayana Korutla, Prashanth Vallabhajosyula, Michael J Campbell, Aisha Khan, Eric Naioti, Keyvan Yousefi, Danial Mehranfard, Lisa McClain-Moss, Anthony A Oliva, Michael E Davis

Abstract

Aims: Hypoplastic left heart syndrome (HLHS) survival relies on surgical reconstruction of the right ventricle (RV) to provide systemic circulation. This substantially increases the RV load, wall stress, maladaptive remodelling, and dysfunction, which in turn increases the risk of death or transplantation.

Methods and results: We conducted a phase 1 open-label multicentre trial to assess the safety and feasibility of Lomecel-B as an adjunct to second-stage HLHS surgical palliation. Lomecel-B, an investigational cell therapy consisting of allogeneic medicinal signalling cells (MSCs), was delivered via intramyocardial injections. The primary endpoint was safety, and measures of RV function for potential efficacy were obtained. Ten patients were treated. None experienced major adverse cardiac events. All were alive and transplant-free at 1-year post-treatment, and experienced growth comparable to healthy historical data. Cardiac magnetic resonance imaging (CMR) suggested improved tricuspid regurgitant fraction (TR RF) via qualitative rater assessment, and via significant quantitative improvements from baseline at 6 and 12 months post-treatment (P < 0.05). Global longitudinal strain (GLS) and RV ejection fraction (EF) showed no declines. To understand potential mechanisms of action, circulating exosomes from intramyocardially transplanted MSCs were examined. Computational modelling identified 54 MSC-specific exosome ribonucleic acids (RNAs) corresponding to changes in TR RF, including miR-215-3p, miR-374b-3p, and RNAs related to cell metabolism and MAPK signalling.

Conclusion: Intramyocardially delivered Lomecel-B appears safe in HLHS patients and may favourably affect RV performance. Circulating exosomes of transplanted MSC-specific provide novel insight into bioactivity. Conduct of a controlled phase trial is warranted and is underway.Trial registration number NCT03525418.

Keywords: Hypoplastic left heart syndrome; Lomecel-B; allogeneic medicinal signalling cells; mesenchymal stem cells; mesenchymal stromal cells.

© The Author(s) 2023. Published by Oxford University Press on behalf of the European Society of Cardiology.

Figures

Structured Graphical Abstract
Structured Graphical Abstract
HLHS, hypoplastic left heart syndrome; MSCs, medicinal signalling cells; RV, Right Ventricular; RA, Right Atrium; SVC, Superior Vena Cava; RPA, Right Pulmonary Artery; LPA, Left Pulmonary Artery
Figure 1
Figure 1
Study timeline and study flow, phase I of the ELPIS study. (A), Study timeline. (B), Study flow. Patients who met the eligibility criteria underwent BDCPA and intramyocardial injections, then were followed up with CMR at 6 and 12 months.
Figure 2
Figure 2
Cardiac function at baseline, 6 months follow-up, and 12-month follow-up in Lomecel-B treated patients (n = 10). (A), Right ventricular ejection fraction did not change over the follow-up period compared to baseline. (B), Right ventricle stroke volume index did not change over the follow-up period compared to baseline. (C), Right ventricle diastolic diameter did not change over the follow-up period compared to baseline. (D), Right ventricle systolic diameter did not change over the follow-up period compared to baseline. (E), Global longitudinal strain did not change over the follow-up period compared to baseline. (F), Tricuspid regurgitation appeared to decrease over the follow-up period compared to baseline.
Figure 3
Figure 3
Tricuspid regurgitation (TR) at baseline, 6-month, and 12-month. (A), TR was assessed by CMR-reader interpretation as mild, moderate, or severe Lomecel-B treated patients (n = 10). (B), TR regurgitant fraction (TR RF) was quantitatively measured from CMR for combined run-in and phase 1 patients. Reported as mean +/− standard deviation. * Indicates P < 0.05.
Figure 4
Figure 4
Baseline and follow-up data monitoring patients throughout ELPIS phase I trial Lomecel-B treated patients (n = 10). (A-B), Serum BNP increased following day two post-treatment but decreased following week twenty-four post-treatment. No changes were statistically significant. Error bars indicate standard deviation. (C), Somatic growth of patients at baseline and at follow-up. Weight for age z-score increased from baseline to 6 months. Weight for age z-score did not differ between baseline and 6 or 12 months. (D), Length for age z-score increased from baseline to 6 months. The length for age z-score significantly decreased from 6-month to 12-months (P = 0.0136) (E), HLA class I reactions significantly decreased from baseline to 26 weeks post-treatment. (F), HLA class II reactions showed a trend towards decreasing after treatment, but this did not reach statistical significance. Only patients with HLA data at baseline and 26 weeks are represented (n = 8). * Indicates P < 0.05. BNP, brain natriuretic peptide.
Figure 5
Figure 5
Unsupervised analysis of MSC-derived exosome RNA. (A), Plasma exosomes from days 2 and 7 were assessed for miRNA and total RNA content with microarray. RNA expression was related to CMR 6 months-to-1 year fold change values. (B), Volcano plot of RNA expression fold change from days 2 to 7. Few miRNAs (circles) and RNAs (squares) upregulated in days 2 and 7 are highlighted in orange and blue, respectively. Principal component analysis of days 2 and 7 miRNAs (C), and RNAs (D). Merged dynamic clustering from weighted correlation network analysis (WGCNA) detected 25 modules of highly correlated day 7 miRNAs (E) and RNAs (F). Module correlation to CMR outcomes (6 months-to-1 year fold change) are depicted in heatmaps (red = high, positive correlation). Modules significantly correlated to an CMR outcome (P < 0.1) are marked. (G), Enriched gene ontology (GO) parent terms of WGCNA RNA modules negatively correlated to TR RF (M2 and M4) and gene targets of miRNA modules positively correlated to TR RF (M12, M20, M24). (H), A network of experimentally validated gene targets of miRNAs positively correlated to TR RF (M12, M20, M24). Enriched terms are coloured by cluster (Metascape).
Figure 6
Figure 6
Partial least-squares regression (PLSR) modelling of day 7 exosome RNA and identification of RNA signals related to TR RF. A two-component model was trained using the top 200 RNA variables of importance for the model projection (VIPs): R2(RNA) = 0.927, R2(CMR Outcomes) = 0.865. (A), Total number of RNAs in the model was reduced to 200 from the initial set of 16 351. A higher proportion of the VIP RNAs are miRNAs. (B), Scores plot of components 1 and 2 from the PLSR model trained with day 7 top 200 VIP RNAs. Components 1 and 2 explain 80% and 13% of the variance, respectively. (C), Loadings plots of components 1 and 2 from the PLSR model show the VIP RNAs covarying with CMR outcomes. (D), PLSR model predictions of CMR outcomes correlated with overserved measurements. RNAs related to TR RF were determined from WGCNA and PLSR analyses. Pearson correlation P-values for RVEF, RV SV, RV Mass, GLS, and TR RF were P < 0.0001, P = 0.0067, P = 0.0005, P = 0.0881, and P = 0.0021 respectively. (E), Venn diagram of WGCNA modules significantly correlated to TR RF (P < 0.001, miRNA M12, M20, M25 and RNA M1, M5) and PLSR 200 VIP RNAs. 54 TR RF-related RNAs identified from the intersection. (F), Top 10 positive and negative weighted coefficients from TR RF-related RNAs were determined with PLSR. (G), Pathway analysis of 54 TR RF-related RNAs (Metascape).

References

    1. Gutgesell HP, Massaro TA. Management of hypoplastic left heart syndrome in a consortium of university hospitals. Am J Cardiol 1995;76:809–811.
    1. Gordon BM, Rodriguez S, Lee M, Chang RK. Decreasing number of deaths of infants with hypoplastic left heart syndrome. J Pediatr 2008;153:354–358.
    1. Reller MD, Strickland MJ, Riehle-Colarusso T, Mahle WT, Correa A. Prevalence of congenital heart defects in metropolitan Atlanta, 1998–2005. J Pediatr 2008;153:807–813.
    1. Hoffman JI, Kaplan S. The incidence of congenital heart disease. J Am Coll Cardiol 2002;39:1890–1900.
    1. Fruitman DS. Hypoplastic left heart syndrome: prognosis and management options. Paediatr Child Health 2000;5:219–225.
    1. Yabrodi M, Mastropietro CW. Hypoplastic left heart syndrome: from comfort care to long-term survival. Pediatr Res 2017;81:142–149.
    1. McGuirk SP, Griselli M, Stumper OF, Rumball EM, Miller P, Dhillon R, de Giovanni JV, Wright JG, Barron DJ, Brawn WJ. Staged surgical management of hypoplastic left heart syndrome: a single institution 12 year experience. Heart 2006;92:364–370.
    1. Feinstein JA, Benson DW, Dubin AM, Cohen MS, Maxey DM, Mahle WT, Pahl E, Villafane J, Bhatt AB, Peng LF, Johnson BA, Marsden AL, Daniels CJ, Rudd NA, Caldarone CA, Mussatto KA, Morales DL, Ivy DD, Gaynor JW, Tweddell JS, Deal BJ, Furck AK, Rosenthal GL, Ohye RG, Ghanayem NS, Cheatham JP, Tworetzky W, Martin GR. Hypoplastic left heart syndrome: current considerations and expectations. J Am Coll Cardiol 2012;59:S1–42.
    1. Best KE, Miller N, Draper E, Tucker D, Luyt K, Rankin J. The improved prognosis of hypoplastic left heart: A population-based register study of 343 cases in England and Wales. Front Pediatr 2021;9:635776.
    1. Everitt MD, Boyle GJ, Schechtman KB, Zheng J, Bullock EA, Kaza AK, Dipchand AI, Naftel DC, Kirklin JK, Canter CE. Pediatric Heart Transplant Study I. Early survival after heart transplant in young infants is lowest after failed single-ventricle palliation: a multi-institutional study. J Heart Lung Transplant 2012;31:509–516.
    1. Williams AR, Trachtenberg B, Velazquez DL, McNiece I, Altman P, Rouy D, Mendizabal AM, Pattany PM, Lopera GA, Fishman J, Zambrano JP, Heldman AW, Hare JM. Intramyocardial stem cell injection in patients with ischemic cardiomyopathy: functional recovery and reverse remodeling. Circ Res 2011;108:792–796.
    1. Hare JM, Fishman JE, Gerstenblith G, DiFede Velazquez DL, Zambrano JP, Suncion VY, Tracy M, Ghersin E, Johnston PV, Brinker JA, Breton E, Davis-Sproul J, Schulman IH, Byrnes J, Mendizabal AM, Lowery MH, Rouy D, Altman P, Wong Po Foo C, Ruiz P, Amador A, Da Silva J, McNiece IK, Heldman AW, George R, Lardo A. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA 2012;308:2369–2379.
    1. Malliaras K, Makkar RR, Smith RR, Cheng K, Wu E, Bonow RO, Marban L, Mendizabal A, Cingolani E, Johnston PV, Gerstenblith G, Schuleri KH, Lardo AC, Marban E. Intracoronary cardiosphere-derived cells after myocardial infarction: evidence of therapeutic regeneration in the final 1-year results of the CADUCEUS trial (CArdiosphere-derived aUtologous stem CElls to reverse ventricUlar dySfunction). J Am Coll Cardiol 2014;63:110–122.
    1. Gunasekaran M, Mishra R, Saha P, Morales D, Cheng WC, Jayaraman AR, Hoffman JR, Davidson L, Chen L, Shah AM, Bittle G, Fu X, Tulshyan A, Abdullah M, Kingsbury T, Civin C, Yang P, Davis ME, Bolli R, Hare JM, Sharma S, Kaushal S. Comparative efficacy and mechanism of action of cardiac progenitor cells after cardiac injury. iScience 2022;25:104656.
    1. Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, Gerstenblith G, DeMaria AN, Denktas AE, Gammon RS, Hermiller JB Jr, Reisman MA, Schaer GL, Sherman W. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol 2009;54:2277–2286.
    1. Nguyen PK, Rhee JW, Wu JC. Adult stem cell therapy and heart failure, 2000 to 2016: A systematic review. JAMA Cardiol 2016;1:831–841.
    1. Banerjee MN, Bolli R, Hare JM. Clinical studies of cell therapy in cardiovascular medicine: recent developments and future directions. Circ Res 2018;123:266–287.
    1. Kanelidis AJ, Premer C, Lopez J, Balkan W, Hare JM. Route of delivery modulates the efficacy of mesenchymal stem cell therapy for myocardial infarction: A meta-analysis of preclinical studies and clinical trials. Circ Res 2017;120:1139–1150.
    1. Wehman B, Sharma S, Pietris N, Mishra R, Siddiqui OT, Bigham G, Li T, Aiello E, Murthi S, Pittenger M, Griffith B, Kaushal S. Mesenchymal stem cells preserve neonatal right ventricular function in a porcine model of pressure overload. Am J Physiol Heart Circ Physiol 2016;310:H1816–H1826.
    1. Premer C, Blum A, Bellio MA, Schulman IH, Hurwitz BE, Parker M, Dermarkarian CR, DiFede DL, Balkan W, Khan A, Hare JM. Allogeneic mesenchymal stem cells restore endothelial function in heart failure by stimulating endothelial progenitor cells. EBioMedicine 2015;2:467–475.
    1. Tarui S, Ishigami S, Ousaka D, Kasahara S, Ohtsuki S, Sano S, Oh H. Transcoronary infusion of cardiac progenitor cells in hypoplastic left heart syndrome: three-year follow-up of the transcoronary infusion of cardiac progenitor cells in patients with single-ventricle physiology (TICAP) trial. J Thorac Cardiovasc Surg 2015;150:1198–1207. 1208 e1–2.
    1. Ishigami S, Ohtsuki S, Eitoku T, Ousaka D, Kondo M, Kurita Y, Hirai K, Fukushima Y, Baba K, Goto T, Horio N, Kobayashi J, Kuroko Y, Kotani Y, Arai S, Iwasaki T, Sato S, Kasahara S, Sano S, Oh H. Intracoronary cardiac progenitor cells in single ventricle physiology: the PERSEUS (cardiac progenitor cell infusion to treat univentricular heart disease) randomized phase 2 trial. Circ Res 2017;120:1162–1173.
    1. Sano T, Ousaka D, Goto T, Ishigami S, Hirai K, Kasahara S, Ohtsuki S, Sano S, Oh H. Impact of cardiac progenitor cells on heart failure and survival in single ventricle congenital heart disease. Circ Res 2018;122:994–1005.
    1. Vincenti M, O’Leary PW, Qureshi MY, Seisler DK, Burkhart HM, Cetta F, Nelson TJ. Clinical impact of autologous cell therapy on hypoplastic left heart syndrome after bidirectional cavopulmonary anastomosis. Semin Thorac Cardiovasc Surg 2021;33:791–801.
    1. Burkhart HM, Qureshi MY, Rossano JW, Cantero Peral S, O’Leary PW, Hathcock M, Kremers W, Nelson TJ. Autologous stem cell therapy for hypoplastic left heart syndrome: safety and feasibility of intraoperative intramyocardial injections. J Thorac Cardiovasc Surg 2019;158:1614–1623.
    1. Ishigami S, Ohtsuki S, Tarui S, Ousaka D, Eitoku T, Kondo M, Okuyama M, Kobayashi J, Baba K, Arai S, Kawabata T, Yoshizumi K, Tateishi A, Kuroko Y, Iwasaki T, Sato S, Kasahara S, Sano S, Oh H. Intracoronary autologous cardiac progenitor cell transfer in patients with hypoplastic left heart syndrome: the TICAP prospective phase 1 controlled trial. Circ Res 2015;116:653–664.
    1. Hare JM, DiFede DL, Rieger AC, Florea V, Landin AM, El-Khorazaty J, Khan A, Mushtaq M, Lowery MH, Byrnes JJ, Hendel RC, Cohen MG, Alfonso CE, Valasaki K, Pujol MV, Golpanian S, Ghersin E, Fishman JE, Pattany P, Gomes SA, Delgado C, Miki R, Abuzeid F, Vidro-Casiano M, Premer C, Medina A, Porras V, Hatzistergos KE, Anderson E, Mendizabal A, Mitrani R, Heldman AW. Randomized comparison of allogeneic versus autologous mesenchymal stem cells for nonischemic dilated cardiomyopathy: POSEIDON-DCM trial. J Am Coll Cardiol 2017;69:526–537.
    1. Kaushal S, Wehman B, Pietris N, Naughton C, Bentzen SM, Bigham G, Mishra R, Sharma S, Vricella L, Everett AD, Deatrick KB, Huang S, Mehta H, Ravekes WA, Hibino N, Difede DL, Khan A, Hare JM. Study design and rationale for ELPIS: A phase I/IIb randomized pilot study of allogeneic human mesenchymal stem cell injection in patients with hypoplastic left heart syndrome. Am Heart J 2017;192:48–56.
    1. Saha P, Sharma S, Korutla L, Datla SR, Shoja-Taheri F, Mishra R, Bigham GE, Sarkar M, Morales D, Bittle G, Gunasekaran M, Ambastha C, Arfat MY, Li D, Habertheuer A, Hu R, Platt MO, Yang P, Davis ME, Vallabhajosyula P, Kaushal S. Circulating exosomes derived from transplanted progenitor cells aid the functional recovery of ischemic myocardium. Sci Transl Med 2019;11:eaau1168.
    1. Pittenger MF, Discher DE, Peault BM, Phinney DG, Hare JM, Caplan AI. Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen Med 2019;4:22.
    1. Andrzejewska A, Lukomska B, Janowski M. Concise review: mesenchymal stem cells: from roots to boost. Stem Cells 2019;37:855–864.
    1. Sotiropoulou PA, Perez SA, Gritzapis AD, Baxevanis CN, Papamichail M. Interactions between human mesenchymal stem cells and natural killer cells. Stem Cells 2006;24:74–85.
    1. Bartholomew A, Sturgeon C, Siatskas M, Ferrer K, McIntosh K, Patil S, Hardy W, Devine S, Ucker D, Deans R, Moseley A, Hoffman R. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol 2002;30:42–48.
    1. Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, Grisanti S, Gianni AM. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 2002;99:3838–3843.
    1. Jiang XX, Zhang Y, Liu B, Zhang SX, Wu Y, Yu XD, Mao N. Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood 2005;105:4120–4126.
    1. Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringden O. HLA Expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol 2003;31:890–896.
    1. Vincenti M, Qureshi MY, Niaz T, Seisler DK, Nelson TJ, Cetta F. Loss of ventricular function after bidirectional cavopulmonary connection: who is at risk? Pediatr Cardiol 2020;41:1714–1724.
    1. Carrillo SA, Texter KM, Phelps C, Tan Y, McConnell PI, Galantowicz M. Tricuspid valve and right ventricular function throughout the hybrid palliation strategy for hypoplastic left heart syndrome and variants. World J Pediatr Congenit Heart Surg 2021;12:9–16.
    1. Borrelli N, Di Salvo G, Sabatino J, Ibrahim A, Avesani M, Sirico D, Josen M, Penco M, Fraisse A, Michielon G. Serial changes in longitudinal strain are associated with outcome in children with hypoplastic left heart syndrome. Int J Cardiol 2020;317:56–62.
    1. Wong J, Lamata P, Rathod RH, Bertaud S, Dedieu N, Bellsham-Revell H, Pushparajah K, Razavi R, Hussain T, Schaeffter T, Powell AJ, Geva T, Greil GF. Right ventricular morphology and function following stage I palliation with a modified Blalock-Taussig shunt versus a right ventricle-to-pulmonary artery conduit. Eur J Cardiothorac Surg 2017;51:50–57.
    1. Lowenthal A, Camacho BV, Lowenthal S, Natal-Hernandez L, Liszewski W, Hills NK, Fineman JR, Bernstein HS. Usefulness of B-type natriuretic peptide and N-terminal pro-B-type natriuretic peptide as biomarkers for heart failure in young children with single ventricle congenital heart disease. Am J Cardiol 2012;109:866–872.
    1. Shah A, Feraco AM, Harmon C, Tacy T, Fineman JR, Bernstein HS. Usefulness of various plasma biomarkers for diagnosis of heart failure in children with single ventricle physiology. Am J Cardiol 2009;104:1280–1284.

Source: PubMed

3
Prenumerera