CAR T cells produced in vivo to treat cardiac injury

Joel G Rurik, István Tombácz, Amir Yadegari, Pedro O Méndez Fernández, Swapnil V Shewale, Li Li, Toru Kimura, Ousamah Younoss Soliman, Tyler E Papp, Ying K Tam, Barbara L Mui, Steven M Albelda, Ellen Puré, Carl H June, Haig Aghajanian, Drew Weissman, Hamideh Parhiz, Jonathan A Epstein, Joel G Rurik, István Tombácz, Amir Yadegari, Pedro O Méndez Fernández, Swapnil V Shewale, Li Li, Toru Kimura, Ousamah Younoss Soliman, Tyler E Papp, Ying K Tam, Barbara L Mui, Steven M Albelda, Ellen Puré, Carl H June, Haig Aghajanian, Drew Weissman, Hamideh Parhiz, Jonathan A Epstein

Abstract

Fibrosis affects millions of people with cardiac disease. We developed a therapeutic approach to generate transient antifibrotic chimeric antigen receptor (CAR) T cells in vivo by delivering modified messenger RNA (mRNA) in T cell–targeted lipid nanoparticles (LNPs). The efficacy of these in vivo–reprogrammed CAR T cells was evaluated by injecting CD5-targeted LNPs into a mouse model of heart failure. Efficient delivery of modified mRNA encoding the CAR to T lymphocytes was observed, which produced transient, effective CAR T cells in vivo. Antifibrotic CAR T cells exhibited trogocytosis and retained the target antigen as they accumulated in the spleen. Treatment with modified mRNA-targeted LNPs reduced fibrosis and restored cardiac function after injury. In vivo generation of CAR T cells may hold promise as a therapeutic platform to treat various diseases.

Conflict of interest statement

Competing Interests: S.M.A., E.P., C.H.J., H.A., D.W., H.P., and J.A.E. are scientific founders and hold equity in Capstan Therapeutics. Y.K.T. and B.L.M. are employees and hold equity in Acuitas Therapeutics. S.M.A. is on the scientific advisory boards of Verismo and Bioardis. C.H.J. is a scientific founder and has equity in Tmunity Therapeutics and DeCART Therapeutics and reports grants from Tmunity Therapeutics and is on the scientific advisory boards of BluesphereBio, Cabaletta, Carisma, Cellares, Celldex, ImmuneSensor, Poseida, Verismo, Viracta Therapeutics, WIRB Copernicus Group and Ziopharm Oncology. D.W. receives research support from BioNTech. S.M.A., E.P., and C.H.J. are inventors (University of Pennsylvania, Wistar Institute) on a patent for a FAP CAR (US Utility Patent 9,365,641 issued 14 June 2016, WIPO Patent Application PCT/US2013/062717). S.M.A., E.P., H.A., and J.A.E. are inventors (University of Pennsylvania) on a patent for the use of CAR T therapy in heart disease (US Provisional Patent Application 62/563,323 filed 26 September 2017, WIPO Patent Application PCT/US2018/052605). J.G.R., I.T., H.A., D.W., H.P., and J.A.E. are inventors (University of Pennsylvania) on a patent for the use of CD5/LNP-FAPCAR as an anti-fibrotic therapy (US Provisional Patent Application 63/090,998 filed 13 September 2020, WIPO Patent Application PCT/ US21/54764 filed 13 October 2021). I.T., D.W., and H.P. are inventors (University of Pennsylvania) on a patent for the in vivo targeting of T cells for mRNA therapeutics (US Provisional Patent Application 63/090,985 filed 13 October 2020, WIPO Patent Application PCT/US21/54769 filed 13 October 2021). I.T., D.W., and H.P. are inventors (University of Pennsylvania) on a patent for the in vivo targeting of CD4+ T cells for mRNA therapeutics (US Provisional Patent Application 63/091,010 filed 13 October 2020, WIPO Patent Application PCT/US21/54775). In accordance with the University of Pennsylvania policies and procedures and our ethical obligations as researchers, D.W. is named on additional patents that describe the use of nucleoside-modified mRNA and targeted lipid nanoparticles as platforms to deliver therapeutic proteins and vaccines. C.H.J. is named on additional patents that describe the creation and therapeutic use of chimeric antigen receptors. These interests have been fully disclosed to the University of Pennsylvania, and approved plans are in place for managing any potential conflicts arising from licensing these patents.

Figures

Fig. 1.. CD5-targeted lipid nanoparticles produce functional,…
Fig. 1.. CD5-targeted lipid nanoparticles produce functional, mRNA-based FAPCAR T cells in vitro.
(A) Schematic outlining the molecular process to create transient FAPCAR T cells using CD5-targeted LNP. Representative flow cytometry analysis of (B) GFP and (C) FAPCAR expression in murine T cells 48 hours after incubation with either IgG/LNP-FAPCAR, CD5/LNP-GFP, or CD5/LNP-FAPCAR. (D) Quantification of murine T cells (percent) staining positive for FAPCAR from biologically independent replicates (n = 4). (E) FAPCAR T cells were mixed with FAP-expressing target HEK293T cells overnight and assayed for killing efficiency in biologically independent replicates (n = 3). Data are mean +/− s.e.m.
Fig. 2.. CD5-targeted lipid nanoparticles produce mRNA-based…
Fig. 2.. CD5-targeted lipid nanoparticles produce mRNA-based FAPCAR T cells in vivo.
(A) Luciferase activity in CD3+ splenocytes 24 hours after intravenous injection of 8µg of control IgG/LNP-Luc or CD5/LNP-Luc. Bar graphs represent two biologically independent replicates. (B) Ai6 mice (Rosa26CAG-LSL-ZsGreen) were injected with 30µg of non-targeted/LNP-Cre (NT), IgG/LNP-Cre, or CD5/LNP-Cre. After 24 hours ZsGreen expression was observed in (81.1%) CD4+ and (75.6%) CD8+, but not in many (15.0%) CD3–splenocytes. Bar graphs represent two biologically independent replicates. (C) T cells were isolated from the spleens of AngII/PE injured mice, 48 hours after injection of 10µg of LNP. Representative flow cytometry analysis shows FAPCAR expression in animals injected with CD5/LNP-FAPCAR, but not in control saline, IgG/LNP-FAPCAR, or CD5/LNP-GFP animals. (D) Quantification of murine T cells staining positive for FAPCAR in C. n = 4 biologically independent mice in two separate cohorts. Data are mean +/− s.e.m.
Fig. 3.. FAPCAR T cells trogocytose FAP…
Fig. 3.. FAPCAR T cells trogocytose FAP from activated cardiac fibroblasts and return FAP to the spleen only in AngII/PE injured, FAPCAR T-treated animals.
(A) Schematic representation of FAPCAR-expressing T cells trogocytosing FAP from activated fibroblasts. (B) Confocal time-lapse micrographs of two FAPCAR T cells first forming an immunological synapse at 40min (arrow), and 85min (arrowhead) then trogocytosing RFP-FAP (magenta) from HEK293T cells (punctae seen at 85min, arrow, and 150min, arrowhead within FAPCAR T cells). Scale bars: 10μm. (C) Widefield images of FAP-stained spleens (white pulp regions highlighted by the dashed line) of an uninjured animal 24 hours after adoptive transfer of 107 MigR1-control T cells, an uninjured animal 24 hours after adoptive transfer of 107 FAPCAR-GFP T cells, an AngII/PE-injured (7 days) animal 48 hours after adoptive transfer of 107 MigR1-control T cells, and an AngII/PE-injured (7 days) animal 48 hours after adoptive transfer of 107 FAPCAR-GFP T cells. Scale bars: 100μm. (D) Confocal micrograph of FAP (magenta) and FAPCAR-GFP (yellow) in a white pulp region of the spleen of an AngII/PE-injured (7 days) animal 48 hours after adoptive transfer of 107 FAPCAR-GFP T cells. Max-Z projection (lower left subpanel) and a single Z slice (lower right subpanel) of a representative FAP+/FAPCAR+ T cell. Scale bars: 10μm. (E) Confocal micrographs of a white pulp region (dashed outline) of FAP-stained spleens from AngII/PE-injured (7 days) animals injected with 10μg of IgG/LNP-FAPCAR or CD5/LNP-FAPCAR for 48 hours. FAP (grey and magenta) and CD3 (yellow) overlap specifically in CD5/LNP-FAPCAR-treated condition. Scale bars: 100μm (top row; greyscale) or 10μm (bottom row; merged pseudo-colored).
Fig. 4.. In vivo generation of transient…
Fig. 4.. In vivo generation of transient FAPCAR T cells improves cardiac function after injury.
Wild-type adult C57BL/6 mice were continually dosed with saline or AngII/PE via implanted 28-day osmotic minipump. After one week of cardiac pressure-overload injury, CD5-targeted LNP were injected. Mice were analyzed after an additional two weeks. (A) Schematic representation of experimental timeline. Echocardiograph measurements show improvements in left ventricle (LV) volumes, diastolic and systolic function following a single injection of 10μg of CD5/LNP-FAPCAR. Measurements of (B) end diastolic and (C) end systolic volumes (μL). (D) M-mode estimate of weight-normalized LV mass (mg/g). (E) Diastolic function (E/e’, an estimate of LV filling pressure) (F) ejection fraction (%) and (G) global longitudinal strain. (H) Representative m-mode echocardiography images. Echocardiograph data represent n = 7, 7, 8 biologically independent mice per condition, spread over three cohorts. (I) Picrosirius red (PSR) staining highlights collagen (pink) in coronal cardiac sections of mock uninjured animals (3 weeks after saline pump implant + saline injection at week 1), injured control animals (AngII/PE + saline), isotype non-targeted LNP control (AngII/PE + IgG/LNP-FAPCAR) and treated animals (AngII/PE + CD5/LNP-FAPCAR). Inset shows magnification of left ventricular myocardium. Scale bar: 100μm. (J) Quantification of percent fibrosis of the ventricles seen in (I). Histology data represent n = 8, 11, 12, biologically independent mice per condition, spread over five cohorts. Data are mean +/− s.e.m. Displayed p-values are from Tukey’s post-hoc test following one-way ANOVA p<0.05.

References

    1. Khalil H, Kanisicak O, Prasad V, Correll RN, Fu X, Schips T, Vagnozzi RJ, Liu R, Huynh T, Lee SJ, Karch J, Molkentin JD, Fibroblast-specific TGF-β-Smad2/3 signaling underlies cardiac fibrosis. J Clin Invest 127, 3770–3783 (2017).
    1. Schafer S, Viswanathan S, Widjaja AA, Lim W-W, Moreno-Moral A, DeLaughter DM, Ng B, Patone G, Chow K, Khin E, Tan J, Chothani SP, Ye L, Rackham OJL, Ko NSJ, Sahib NE, Pua CJ, Zhen NTG, Xie C, Wang M, Maatz H, Lim S, Saar K, Blachut S, Petretto E, Schmidt S, Putoczki T, Guimarães-Camboa N, Wakimoto H, van Heesch S, Sigmundsson K, Lim SL, Soon JL, Chao VTT, Chua YL, Tan TE, Evans SM, Loh YJ, Jamal MH, Ong KK, Chua KC, Ong B, Chakaramakkil MJ, Seidman JG, Seidman CE, Hubner N, Sin KYK, Cook SA, IL-11 is a crucial determinant of cardiovascular fibrosis. Nature 552, 110–115 (2017).
    1. Moore-Morris T, Guimarães-Camboa N, Banerjee I, Zambon AC, Kisseleva T, Velayoudon A, Stallcup WB, Gu Y, Dalton ND, Cedenilla M, Gomez-Amaro R, Zhou B, Brenner DA, Peterson KL, Chen J, Evans SM, Resident fibroblast lineages mediate pressure overload–induced cardiac fibrosis. J Clin Invest 124, 2921–2934 (2014).
    1. Yokota T, McCourt J, Ma F, Ren S, Li S, Kim TH, Kurmangaliyev YZ, Nasiri R, Ahadian S, Nguyen T, Tan XHM, Zhou Y, Wu R, Rodriguez A, Cohn W, Wang Y, Whitelegge J, Ryazantsev S, Khademhosseini A, Teitell MA, Chiou PY, Birk DE, Rowat AC, Crosbie RH, Pellegrini M, Seldin M, Lusis AJ, Deb A, Type V Collagen in Scar Tissue Regulates the Size of Scar after Heart Injury. Cell 182, 545–562.e23 (2020).
    1. Rurik JG, Aghajanian H, Epstein JA, Immune Cells and Immunotherapy for Cardiac Injury and Repair. Circ Res 128, 1766–1779 (2021).
    1. Widjaja AA, Viswanathan S, Jinrui D, Singh BK, Tan J, Wei Ting JG, Lamb D, Shekeran SG, George BL, Schafer S, Carling D, Adami E, Cook SA, Molecular Dissection of Pro-Fibrotic IL11 Signaling in Cardiac and Pulmonary Fibroblasts. Front Mol Biosci 8, 1–14 (2021).
    1. Henderson NC, Rieder F, Wynn TA, Fibrosis: from mechanisms to medicines. Nature 587, 555–566 (2020).
    1. González A, Schelbert EB, Díez J, Butler J, Myocardial Interstitial Fibrosis in Heart Failure: Biological and Translational Perspectives. J Am Coll Cardiol 71, 1696–1706 (2018).
    1. Aghajanian H, Kimura T, Rurik JG, Hancock AS, Leibowitz MS, Li L, Scholler J, Monslow J, Lo A, Han W, Wang T, Bedi K, Morley MP, Linares Saldana RA, Bolar NA, McDaid K, Assenmacher C, Smith CL, Wirth D, June CH, Margulies KB, Jain R, Puré E, Albelda SM, Epstein JA, Targeting cardiac fibrosis with engineered T cells. Nature 573, 430–433 (2019).
    1. Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, June CH, T Cells with Chimeric Antigen Receptors Have Potent Antitumor Effects and Can Establish Memory in Patients with Advanced Leukemia. Sci Transl Med 3, 95ra73-95ra73 (2011).
    1. Weissman D, mRNA transcript therapy. Expert Rev Vaccines 14, 265–281 (2014).
    1. Karikó K, Buckstein M, Ni H, Weissman D, Suppression of RNA recognition by Toll-like receptors: The impact of nucleoside modification and the evolutionary origin of RNA. Immunity 23, 165–175 (2005).
    1. Karikó K, Muramatsu H, Welsh FA, Ludwig J, Kato H, Akira S, Weissman D, Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stability. Mol Ther 16, 1833–1840 (2008).
    1. Andries O, Mc Cafferty S, De Smedt SC, Weiss R, Sanders NN, Kitada T, N1-methylpseudouridine-incorporated mRNA outperforms pseudouridine-incorporated mRNA by providing enhanced protein expression and reduced immunogenicity in mammalian cell lines and mice. J Control Release 217, 337–344 (2015).
    1. Zhao Y, Moon E, Carpenito C, Paulos CM, Liu X, Brennan AL, Chew A, Carroll RG, Scholler J, Levine BL, Albelda SM, June CH, Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor. Cancer Res 70, 9053–9061 (2010).
    1. Pardi N, Hogan MJ, Porter FW, Weissman D, mRNA vaccines-a new era in vaccinology. Nat Rev Drug Discov 17, 261–279 (2018).
    1. Rizvi F, Everton E, Smith AR, Liu H, Osota E, Beattie M, Tam Y, Pardi N, Weissman D, Gouon-Evans V, Murine liver repair via transient activation of regenerative pathways in hepatocytes using lipid nanoparticle-complexed nucleoside-modified mRNA. Nat Commun 12 (2021), doi:10.1038/s41467-021-20903-3.
    1. Krienke C, Kolb L, Diken E, Streuber M, Kirchhoff S, Bukur T, Akilli-Öztürk Ö, Kranz LM, Berger H, Petschenka J, Diken M, Kreiter S, Yogev N, Waisman A, Karikó K, Türeci Ö, Sahin U, A noninflammatory mRNA vaccine for treatment of experimental autoimmune encephalomyelitis. Science (80-). 371, 145–153 (2021).
    1. Szőke D, Kovács G, Kemecsei É, Bálint L, Szoták-Ajtay K, Aradi P, Styevkóné Dinnyés A, Mui BL, Tam YK, Madden TD, Karikó K, Kataru RP, Hope MJ, Weissman D, Mehrara BJ, Pardi N, Jakus Z, Nucleoside-modified VEGFC mRNA induces organ-specific lymphatic growth and reverses experimental lymphedema. Nat Commun 12, 3460 (2021).
    1. Gillmore JD, Gane E, Taubel J, Kao J, Fontana M, Maitland ML, Seitzer J, O’Connell D, Walsh KR, Wood K, Phillips J, Xu Y, Amaral A, Boyd AP, Cehelsky JE, McKee MD, Schiermeier A, Harari O, Murphy A, Kyratsous CA, Zambrowicz B, Soltys R, Gutstein DE, Leonard J, Sepp-Lorenzino L, Lebwohl D, CRISPR-Cas9 In Vivo Gene Editing for Transthyretin Amyloidosis. N Engl J Med, 1–10 (2021).
    1. Pardi N, Tuyishime S, Muramatsu H, Kariko K, Mui BL, Tam YK, Madden TD, Hope MJ, Weissman D, Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes. J Control Release 217, 345–351 (2015).
    1. Akinc A, Querbes W, De S, Qin J, Frank-Kamenetsky M, Jayaprakash KN, Jayaraman M, Rajeev KG, Cantley WL, Dorkin JR, Butler JS, Qin L, Racie T, Sprague A, Fava E, Zeigerer A, Hope MJ, Zerial M, Sah DW, Fitzgerald K, Tracy MA, Manoharan M, Koteliansky V, De Fougerolles A, Maier MA, Targeted delivery of RNAi therapeutics with endogenous and exogenous ligand-based mechanisms. Mol Ther 18, 1357–1364 (2010).
    1. Parhiz H, Shuvaev VV, Pardi N, Khoshnejad M, Kiseleva RY, Brenner JS, Uhler T, Tuyishime S, Mui BL, Tam YK, Madden TD, Hope MJ, Weissman D, Muzykantov VR, PECAM-1 directed re-targeting of exogenous mRNA providing two orders of magnitude enhancement of vascular delivery and expression in lungs independent of apolipoprotein E-mediated uptake. J Control Release 291, 106–115 (2018).
    1. Tombácz I, Laczkó D, Shahnawaz H, Muramatsu H, Natesan A, Yadegari A, Papp TE, Alameh M-G, Shuvaev V, Mui BL, Tam YK, Muzykantov V, Pardi N, Weissman D, Parhiz H, Highly efficient CD4+ T cell targeting and genetic recombination using engineered CD4+ cell-homing mRNA-LNP. Mol Ther (2021), doi:10.1016/j.ymthe.2021.06.004.
    1. Wang LCS, Lo A, Scholler J, Sun J, Majumdar RS, Kapoor V, Antzis M, Cotner CE, Johnson LA, Durham AC, Solomides CC, June CH, Puré E, Albelda SM, Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicity. Cancer Immunol Res 2, 154–166 (2014).
    1. Boumsell L, Coppin H, Pham D, Raynal B, Lemerle J, Dausset J, Bernard A, An antigen shared by a human T cell subset and B cell chronic lymphocytic leukemic cells. Distribution on normal and malignant lymphoid cells. J Exp Med 152, 229–234 (1980).
    1. Soldevila G, Raman C, Lozano F, The immunomodulatory properties of the CD5 lymphocyte receptor in health and disease. Curr Opin Immunol 23, 310–318 (2011).
    1. Kaur H, Takefuji M, Ngai CY, Carvalho J, Bayer J, Wietelmann A, Poetsch A, Hoelper S, Conway SJ, Möllmann H, Looso M, Troidl C, Offermanns S, Wettschureck N, Targeted Ablation of Periostin-Expressing Activated Fibroblasts Prevents Adverse Cardiac Remodeling in Mice. Circ Res 118, 1906–1917 (2016).
    1. Sommermeyer D, Hudecek M, Kosasih PL, Gogishvili T, Maloney DG, Turtle CJ, Riddell SR, Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia 30, 492–500 (2016).
    1. Hamieh M, Dobrin A, Cabriolu A, van der Stegen SJC, Giavridis T, Mansilla-Soto J, Eyquem J, Zhao Z, Whitlock BM, Miele MM, Li Z, Cunanan KM, Huse M, Hendrickson RC, Wang X, Rivière I, Sadelain M, CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape. Nature 568, 112–116 (2019).
    1. Joly E, Hudrisier D, What is trogocytosis and what is its purpose? Nat Immunol 4, 815–815 (2003).
    1. Martínez-Martín N, Fernández-Arenas E, Cemerski S, Delgado P, Turner M, Heuser J, Irvine DJ, Huang B, Bustelo XR, Shaw A, Alarcón B, T Cell Receptor Internalization from the Immunological Synapse Is Mediated by TC21 and RhoG GTPase-Dependent Phagocytosis. Immunity 35, 208–222 (2011).
    1. Newick K, O’brien S, Sun J, Kapoor V, Maceyko S, Lo A, Puré E, Moon E, Albelda SM, Augmentation of CAR T-cell trafficking and antitumor efficacy by blocking protein kinase a localization. Cancer Immunol Res 4, 541–551 (2016).
    1. Baiersdörfer M, Boros G, Muramatsu H, Mahiny A, Vlatkovic I, Sahin U, Karikó K, A Facile Method for the Removal of dsRNA Contaminant from In Vitro-Transcribed mRNA. Mol Ther - Nucleic Acids 15, 26–35 (2019).
    1. Maier MA, Jayaraman M, Matsuda S, Liu J, Barros S, Querbes W, Tam YK, Ansell SM, Kumar V, Qin J, Zhang X, Wang Q, Panesar S, Hutabarat R, Carioto M, Hettinger J, Kandasamy P, Butler D, Rajeev KG, Pang B, Charisse K, Fitzgerald K, Mui BL, Du X, Cullis P, Madden TD, Hope MJ, Manoharan M, Akinc A, Biodegradable lipids enabling rapidly eliminated lipid nanoparticles for systemic delivery of RNAi therapeutics. Mol Ther 21, 1570–1578 (2013).
    1. Pedram A, Razandi M, Lubahn D, Liu J, Vannan M, Levin ER, Estrogen inhibits cardiac hypertrophy: Role of estrogen receptor-β to inhibit calcineurin. Endocrinology 149, 3361–3369 (2008).

Source: PubMed

3
S'abonner