Cardiac Function Improvement and Bone Marrow Response -: Outcome Analysis of the Randomized PERFECT Phase III Clinical Trial of Intramyocardial CD133+ Application After Myocardial Infarction

Gustav Steinhoff, Julia Nesteruk, Markus Wolfien, Günther Kundt, PERFECT Trial Investigators Group, Jochen Börgermann, Robert David, Jens Garbade, Jana Große, Axel Haverich, Holger Hennig, Alexander Kaminski, Joachim Lotz, Friedrich-Wilhelm Mohr, Paula Müller, Robert Oostendorp, Ulrike Ruch, Samir Sarikouch, Anna Skorska, Christof Stamm, Gudrun Tiedemann, Florian Mathias Wagner, Olaf Wolkenhauer, Gustav Steinhoff, Julia Nesteruk, Markus Wolfien, Günther Kundt, PERFECT Trial Investigators Group, Jochen Börgermann, Robert David, Jens Garbade, Jana Große, Axel Haverich, Holger Hennig, Alexander Kaminski, Joachim Lotz, Friedrich-Wilhelm Mohr, Paula Müller, Robert Oostendorp, Ulrike Ruch, Samir Sarikouch, Anna Skorska, Christof Stamm, Gudrun Tiedemann, Florian Mathias Wagner, Olaf Wolkenhauer

Abstract

Objective: The phase III clinical trial PERFECT was designed to assess clinical safety and efficacy of intramyocardial CD133+ bone marrow stem cell treatment combined with CABG for induction of cardiac repair.

Design: Multicentre, double-blinded, randomised placebo controlled trial.

Setting: The study was conducted across six centres in Germany October 2009 through March 2016 and stopped due slow recruitment after positive interim analysis in March 2015.

Participants: Post-infarction patients with chronic ischemia and reduced LVEF (25-50%).

Interventions: Eighty-two patients were randomised to two groups receiving intramyocardial application of 5ml placebo or a suspension of 0.5-5×106 CD133+.

Outcome: Primary endpoint was delta (∆) LVEF at 180days (d) compared to baseline measured in MRI.

Findings (prespecified): Safety (n=77): 180d survival was 100%, MACE n=2, SAE n=49, without difference between placebo and CD133+. Efficacy (n=58): The LVEF improved from baseline LVEF 33.5% by +9.6% at 180d, p=0.001 (n=58). Treatment groups were not different in ∆LVEF (ANCOVA: Placebo +8.8% vs. CD133+ +10.4%, ∆CD133+vs placebo +2.6%, p=0.4).

Findings (post hoc): Responders (R) classified by ∆LVEF≥5% after 180d were 60% of the patients (35/58) in both treatment groups. ∆LVEF in ANCOVA was +17.1% in (R) vs. non-responders (NR) (∆LVEF 0%, n=23). NR were characterized by a preoperative response signature in peripheral blood with reduced CD133+ EPC (RvsNR: p=0.005) and thrombocytes (p=0.004) in contrast to increased Erythropoeitin (p=0.02), and SH2B3 mRNA expression (p=0.073). Actuarial computed mean survival time was 76.9±3.32months (R) vs. +72.3±5.0months (NR), HR 0.3 [Cl 0.07-1.2]; p=0.067.Using a machine learning 20 biomarker response parameters were identified allowing preoperative discrimination with an accuracy of 80% (R) and 84% (NR) after 10-fold cross-validation.

Interpretation: The PERFECT trial analysis demonstrates that the regulation of induced cardiac repair is linked to the circulating pool of CD133+ EPC and thrombocytes, associated with SH2B3 gene expression. Based on these findings, responders to cardiac functional improvement may be identified by a peripheral blood biomarker signature.

Trial registration: ClinicalTrials.govNCT00950274.

Keywords: Angiogenesis; CD133(+); CD34(+); Cardiac repair; Cardiac stem cell therapy; Endothelial progenitor cell (EPC); Lnk adaptor; Randomised double-blinded phase III multicentre trial; SH2B3.

Copyright © 2017 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Fig. 1
Fig. 1
PERFECT Trial flowchart and prespecified or post hoc analysis sets. The randomised multicentre trial was performed double-blinded placebo controlled through six heart centres in Germany according to ICH-GCP and is depicted according to CONSORT and STARD guidelines:1 A total of 119 patients were screened in 6 centres in Germany from Sept. 2009 through June 2015. All patients signed the informed consent form and were included in the study. Thirty-seven participants were excluded before randomisation due to newly identified exclusion criteria such as severe arrhythmia. 2 Eighty-two (82) patients were randomised to active treatment or placebo. Two (Stamm et al., 2003) patients were randomised but not treated because the CD 133 + preparation did not comply with the release criteria for GMP. 3 Forty (48.8%) patients received an injection of CD133 + cells and 40 (48 ⋅ 8%) received an injection of placebo. 4 Three patients were excluded because of insufficient CD133 + cell count below minimum dosis resulting in the safety-analysis-population (n = 77). 5 After a careful review of the blinded data in a blind data review meeting conducted on the 20 May 2016 a total of 19 patients were excluded from the full analysis population due to protocol violations with incomplete MRI follow-up data leading to the Per Protocol Set (PPS) efficacy-analysis-population (n = 58). Patient distribution for PPS efficacy population by study centres: German Heart Center Berlin 8%, Medical School Hannover 28%, University Medicine Rostock 38%, Heart and Diabetes Center Bad Oeynhausen 5%, Heart Center Leipzig 13%, University Medicine Hamburg 10%. 6 Additional MRI at day 10 postoperative for subanalysis of early and late postoperative changes for subgroup analysis early and late postoperative changes. 7 Post hoc analysis for actuarial survival was performed in registry analysis 7 years after FPI on Nov. 1, 2016. 8 Post hoc analysis was additionally performed in the efficacy group (n = 58) to unravel contributing non CD133+ injection related factors of late improvement. Patients were grouped in the efficacy analysis set according to effective response in primary endpoint as responder or non-responder (Δ LVEF 180 d vs.0 responder ≥ 5% vs. non-responder < 5%). According to this post hoc analysis 35 patients from 58 (60.3%) were responders to treatment. This Responder/non-responder (R/NR) ratio was similar respectively in the placebo group 56 ⋅ 5% (R/NR 17/30 pt.) and in the CD133 + group 64% (R/NR 18/28 pt.) (Placebo vs. CD133 +; p = 0 ⋅ 373). Responder (35/58) and non-responder (23/58) analysis was performed in efficacy group (n = 58). 9 Biomarkers were studied in 39 patients of the efficacy group (n = 58) independent on placebo/CD133 + or responder/non-responder group. All laboratory tests were realized in patients located in the Rostock centre (n = 31), where immediate laboratory analysis of FACS and CFU was guaranteed. Additional patients from other centres (8/58) were evaluated also in the Biomarker cohort according to realized parameters. Biobank at time point (pre- and postoperative day) − 2, − 1, + 1, + 3, + 10, + 180: Peripheral blood MNC/FACS (CD 133, 34, 117, 184, 309, 45, 31, 14), CFU-Hill, serum analysis angiogenesis factors and cytokines; Bone-marrow MNC, Isolated CD133 + FACS (CD133, 34, 117, 184, 309, 45, 31, 14), CFU-EC, RNA-seq.
Fig. 2
Fig. 2
Early and late recovery of LVEF in Placebo and CD133+ groups. MRI analysis of LVEF (%) is depicted in 29 patients with intermediate MRI at day 10 postoperatively and at 180 days. *p value for delta LVEF at 10 days versus 0. #p value for delta LVEF at 6 months versus 10 days.
Fig. 3
Fig. 3
a: Kaplan-Meier survival analysis in longterm follow-up: Placebo vs. CD133+. b: Kaplan-Meier survival analysis in longterm follow-up: Responder vs. Non-responder
Fig. 4
Fig. 4
SH2B3 expression analysis in peripheral blood of responder and non-responder. Whole blood samples were obtained from 21 patients before coronary artery bypass graft (CABG) revascularization. Relative expression of SH2B3 (a) and corresponding ΔCT values (b) were calculated using the 2− ΔΔCT method. All values are presented as mean ± SEM and normalized to GAPDH and POLR2A. n = 13 (responder); n = 8 (non-responder). ΔCT values: p = 0.073.
Fig. 5
Fig. 5
a Three-dimensional t-SNE calculation of the Rostock subgroup. The variables x and y refer to the newly calculated features that are used to classify the patients into distinct groups. The model was subsequently fitted by a polynomial (n3) equation to visualize the z-axis as a geographic profile. The respective colors for the responder (red dot) and non-responder (grey dot) patients have been added afterwards. The classified groups have been roughly summarized by a red and grey dashed line. Results are obtained after 3000 iterations. The calculation of the ratio between responder and non-responder is indicated for each circle. It is more likely for the non-responder group to be located at smaller z-values (z < 20, ratio < 42%). The responders tend to be enriched within the light blue areas (z > 20) including a ration > 69%. b Obtained supervised ML prediction results for pre- and postoperative time points (0 days to 180 days) of the clinical and clinical & laboratory dataset to distinguish between responder and non-responder. The graph shows the true positive prediction results of five independent feature selected ML models (AdaBoost for feature selection and RF for final prediction).The error bars indicate the respective accuracy standard deviation for the constructed models that have been obtained after 100 iterations. The 100 model iterations are significant different according to one-way ANOVA (p < 0.001).
Fig. 6
Fig. 6
Outcome results of the PERFECT trial.

References

    1. Auer P.L., Teumer A., Schick U. Rare and low-frequency coding variants in CXCR2 and other genes are associated with hematological traits. Nat. GenetNat. Genet. 2014;46(6):629–634. (Epub 2014 Apr 28)
    1. Bartunek J., Terzic A., Davison B.A. Cardiopoietic cell therapy for advanced ischemic heart failure: results at 39 weeks of the prospective, randomized, double blind, sham-controlled CHART-1 clinical trial. Eur. Heart J. 2016 Dec 23. pii: ehw543. (Epub ahead of print)
    1. Bartunek J., Terzic A., Davison B.A. Cardiopoietic cell therapy for advanced ischaemic heart failure: results at 39 weeks of the prospective, randomized, double blind, sham-controlled CHART-1 clinical trial. Eur. Heart J. 2017;38(9):648–660.
    1. Bhatnagar A., Bolli R., Johnstone B.H. Cardiovascular cell therapy research network (CCTRN). Bone marrow cell characteristics associated with patient profile and cardiac performance outcomes in the LateTIME-cardiovascular cell therapy research network (CCTRN) trial. Am. Heart J. 2016;179:142–150.
    1. Blatt A., Elbaz-Greener G.A., Tuby H., Maltz L. Low-level laser therapy to the bone marrow reduces scarring and improves heart function post-acute myocardial infarction in the pig. Photomed. Laser Surg. 2016;34(11):516–524.
    1. Cesari F., Caporale R., Marcucci R. NT-proBNP and the anti-inflammatory cytokines are correlated with endothelial progenitor cells' response to cardiac surgery. Atherosclerosis. 2008;199(1):138–146.
    1. Contreras A., Orozco A.F., Resende M. Identification of cardiovascular risk factors associated with bone marrow cell subsets in patients with STEMI: a biorepository evaluation from the CCTRN TIME and LateTIME clinical trials. Basic Res. Cardiol. 2017;112(1):3.
    1. Donndorf P., Kaminski A., Tiedemann G., Kundt G., Steinhoff G. Validating intramyocardial bone marrow stem cell therapy in combination with coronary artery bypass grafting, the PERFECT phase III randomized multicenter trial: study protocol for a randomized controlled trial. Trials. 2012;13:99.
    1. Fisher S.A., Mathur A., Taggart D.P., Martin-Rendon E. Stem cell therapy for chronic ischaemic heart disease and congestive heart failure. Cochrane Database Syst. Rev. 2016;12
    1. Forman G., Cohen I. 2004. Learning From Little: Comparison of Classifiers Given Little Training.
    1. Fortney K., Dobriban E., Garagnani P. Genome-wide scan informed by age-related disease identifies loci for exceptional human longevity. PLoS Genet. 2015;11(12):e1005728.
    1. Henry Timothy D., Moyé Lem, Traverse Jay H. Consistently inconsistent—bone marrow mononuclear stem cell therapy following acute myocardial infarction - a decade later. Circ. Res. 2016;119:404–406.
    1. Hofmann W.K., de Vos S., Elashoff D. Relation between resistance of Philadelphia-chromosome-positive acute lymphoblastic leukaemia to the tyrosine kinase inhibitor STI571 and gene-expression profiles: a gene-expression study. Lancet. 2002;359(9305):481–486.
    1. Ishige-Wada M., Kwon S.M., Eguchi M. Jagged-1 signaling in the bone marrow microenvironment promotes endothelial progenitor cell expansion and commitment of CD133 + human cord blood cells for postnatal Vasculogenesis. PLoS One. 2016;11(11):e0166660.
    1. Kuhn M. Building Predictive Models in R using the caret package. J. Stat. Softw. 2008;28(5):1–26.
    1. Kwon S.M., Suzuki T., Kawamoto A., Ii M., Eguchi M., Akimaru H., Wada M., Matsumoto T., Masuda H., Nakagawa Y., Nishimura H., Kawai K., Takaki S., Asahara T. Pivotal role of lnk adaptor protein in endothelial progenitor cell biology for vascular regeneration. Circ. Res. 2009;104(8):969–977.
    1. Lee Jun Hee, Ji Seung Taek, Kim Jaeho. Specific disruption of Lnk in murine endothelial progenitor cells promotes dermal wound healing via enhanced vasculogenesis, activation of myofibroblasts, and suppression of inflammatory cell recruitment. Stem Cell Res Ther. 2016;7(1):158.
    1. Maaten L.V.D., Hinton G. Visualizing data using t-SNE. J. Mach. Learn. Res. 2008;9:2579–2605.
    1. McPherson R., Tybjaerg-Hansen A. Genetics of coronary artery disease. Circ. Res. 2016;118(4):564–578. Feb 19.
    1. Nasseri B.A., Ebell W., Dandel M. Autologous CD133 + bone marrow cells and bypass grafting for regeneration of ischaemic myocardium: the Cardio133 trial. Eur. Heart J. 2014;35(19):1263–1274.
    1. O'Brien R.G., Muller K.E., Edward L.K., editors. Applied Analysis of Variance in Behavioral Science. Marcel Dekker; New York: 1993. Signified power analysis for t-tests through multivariate hypotheses.
    1. Rosenberger William F., Lachin John M. Wiley publ; 2003. Randomization in Clinical Trials: Theory and Practice; pp. 1–14.
    1. Saeb A.T., Al-Naqeb D. The impact of evolutionary driving forces on human complex diseases: a population genetics approach. Scientifica (Cairo) 2016;2016 (Epub 2016 May 30)
    1. Stamm C., Westphal B., Kleine H.D. Autologous bone-marrow stem-cell transplantation for myocardial regeneration. Lancet. 2003;361(9351):45–46.
    1. Stamm C., Kleine H.D., Choi Y.H. Intramyocardial delivery of CD133 + bone marrow cells and coronary artery bypass grafting for chronic ischemic heart disease: safety and efficacy studies. J. Thorac. Cardiovasc. Surg. 2007;133(3):717–725.
    1. Strieter R.M., Kunkel S.L., Arenberg D.A., Burdick M.D., Polverini P.J. Interferon gamma-inducible protein 10 (IP-10), a member of the C-X-C chemokine family, is an inhibitor of angiogenesis. Biochem. Biophys. Res. Commun. 1995;210(1):51–57.
    1. Takizawa H., Eto K., Yoshikawa A., Nakauchi H., Takatsu K., Takaki S. Growth and maturation of megakaryocytes is regulated by Lnk/Sh2b3 adaptor protein through crosstalk between cytokine- and integrin-mediated signals. Exp. Hematol. 2008 Jul;36(7):897–906.
    1. Taylor D.A., Perin E.C., Willerson J.T. Cardiovascular cell therapy research network (CCTRN). Identification of bone marrow cell subpopulations associated with improved functional outcomes in patients with chronic left ventricular dysfunction: an embedded cohort evaluation of the FOCUS-CCTRN trial. Cell Transplant. 2016;25(9):1675–1687.
    1. Tian T., Chen B., Xiao Y., Yang K., Zhou X., Zhou X. Intramyocardial autologous bone marrow cell transplantation for ischemic heart disease: a systematic review and meta-analysis of randomized controlled trials. Atherosclerosis. 2014;233(2):485–492.
    1. Tse H.F., Kwong Y.L., Chan J.K., Lo G., Ho C.L., Lau C.P. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet. 2003;361(9351):47–49.
    1. Vakil K., Florea V., Koene R. Effect of coronary artery bypass grafting on left ventricular ejection fraction in men eligible for implantable cardioverter-defibrillator. Am. J. Cardiol. 2016;117:957–960.
    1. Werner N., Kosiol S., Schiegl T. Circulating endothelial progenitor cells and cardiovascular outcomes. N. Engl. J. Med. 2005;353(10):999–1007.

Source: PubMed

3
Abonneren