Cell penetrating peptide functionalized perfluorocarbon nanoemulsions for targeted cell labeling and enhanced fluorine-19 MRI detection
Dina V. Hingorani
Department of Radiology, University of California San Diego, California
Search for more papers by this authorFanny Chapelin
Department of Bioengineering, University of California San Diego, California
Search for more papers by this authorEmma Stares
Department of Radiology, University of California San Diego, California
Search for more papers by this authorStephen R. Adams
Department of Pharmacology, University of California San Diego, California
Search for more papers by this authorHideho Okada
Department of Neurological Surgery, University of California San Francisco, California
Search for more papers by this authorCorresponding Author
Eric T. Ahrens
Department of Radiology, University of California San Diego, California
Correspondence
Eric T. Ahrens, Department of Radiology, University of California San Diego, 9500 Gilman Dr. #0695, La Jolla, CA 92093-0695.
Email: [email protected]
Search for more papers by this authorDina V. Hingorani
Department of Radiology, University of California San Diego, California
Search for more papers by this authorFanny Chapelin
Department of Bioengineering, University of California San Diego, California
Search for more papers by this authorEmma Stares
Department of Radiology, University of California San Diego, California
Search for more papers by this authorStephen R. Adams
Department of Pharmacology, University of California San Diego, California
Search for more papers by this authorHideho Okada
Department of Neurological Surgery, University of California San Francisco, California
Search for more papers by this authorCorresponding Author
Eric T. Ahrens
Department of Radiology, University of California San Diego, California
Correspondence
Eric T. Ahrens, Department of Radiology, University of California San Diego, 9500 Gilman Dr. #0695, La Jolla, CA 92093-0695.
Email: [email protected]
Search for more papers by this authorAbstract
Purpose
A bottleneck in developing cell therapies for cancer is assaying cell biodistribution, persistence, and survival in vivo. Ex vivo cell labeling using perfluorocarbon (PFC) nanoemulsions, paired with 19F MRI detection, is a non-invasive approach for cell product detection in vivo. Lymphocytes are small and weakly phagocytic limiting PFC labeling levels and MRI sensitivity. To boost labeling, we designed PFC nanoemulsion imaging probes displaying a cell-penetrating peptide, namely the transactivating transcription sequence (TAT) of the human immunodeficiency virus. We report optimized synthesis schemes for preparing TAT co-surfactant to complement the common surfactants used in PFC nanoemulsion preparations.
Methods
We performed ex vivo labeling of primary human chimeric antigen receptor (CAR) T cells with nanoemulsion. Intracellular labeling was validated using electron microscopy and confocal imaging. To detect signal enhancement in vivo, labeled CAR T cells were intra-tumorally injected into mice bearing flank glioma tumors.
Results
By incorporating TAT into the nanoemulsion, a labeling efficiency of ~1012 fluorine atoms per CAR T cell was achieved that is a >8-fold increase compared to nanoemulsion without TAT while retaining high cell viability (~84%). Flow cytometry phenotypic assays show that CAR T cells are unaltered after labeling with TAT nanoemulsion, and in vitro tumor cell killing assays display intact cytotoxic function. The 19F MRI signal detected from TAT-labeled CAR T cells was 8 times higher than cells labeled with PFC without TAT.
Conclusion
The peptide-PFC nanoemulsion synthesis scheme presented can significantly enhance cell labeling and imaging sensitivity and is generalizable for other targeted imaging probes.
CONFLICT OF INTEREST
ETA is founder, consultant, member of the advisory board and shareholder of Celsense, Inc.
Supporting Information
Filename | Description |
---|---|
mrm27988-sup-0001-FigS1-S8.pdfPDF document, 625 KB |
FIGURE S1 Synthesis scheme of F68-TAT co-surfactant. F68 is functionalized with a maleimide group to enable addition of the TAT peptide with a terminal cysteine (Cys-TAT) FIGURE S2 Size stability of TAT-F68-PFC nanoemulsions. The effect of % TAT incorporation on size (A) and polydispersity index (PDI) (B) of nanoemulsions is shown. The nanoemulsion size (C) and PDI (D) of nanoemulsions over time while stored at 4°C is displayed FIGURE S3 Optimization of lipid-TAT-PFC incubation time in Jurkat cells. Incubation times of 2, 4, and 18 h are tested as shown in (A), and the highest uptake is observed at 18 h. Jurkat cell viability is not altered by labeling for different durations (B) FIGURE S4 Cy5-TATA,P-F68-PFC synthesis scheme. Scheme shows synthesis of fluorescently labeled co-surfactants 8 and 9 consisting of Cy5 dye attached to the respective fluorous anchors 6 and 7 for incorporation into TATP-F68-PFC and TATA-F68-PFC nanoemulsions FIGURE S5 Localization impact of incorporation of fluorescent dye into surfactant layer during nanoemulsion preparation. (A) 19F uptake for cells treated with nanoemulsions prepared with and without anchored Cy5 at 10 mg/mL and 20 mg/mL doses; no significant differences are observed. Additionally, CAR T cell viability is not affected as shown in (B). (C) Intracellular localization of the nanoemulsion (Cy5 in red) in CAR T cells via confocal microscopy. Hoechst dye (nuclei, blue) and Alexa488 dye (cell membrane, green) is used to delineate cell structures FIGURE S6 Fluorescent dye conjugate nanoemulsions without TAT do not get internalized into CAR T cells. Panels show that dye compounds 8 and 9 do not induce non-specific internalization into live cells. Hoechst dye (nuclei, blue) and Alexa488 dye (cell membrane, green) are used to delineate the cells FIGURE S7 CAR T cell killing assay in vitro. Co-incubation of human U87-EGFRvIII-Luc glioma cells with TATP-F68-PFC-labeled or unlabeled CAR T cells, or untransduced T cells results in significant cell death at 12 and 24 h. CAR T cells exhibit significant tumor killing ability (~98%) compared to untransduced T cells (~60%). Killing efficacy is unaltered by nanoemulsion labeling of the cells FIGURE S8 Ex vivo 3D microimaging of excised glioma tumors harboring PFC-labeled CAR T cells. Contiguous images show overlays of 19F (pseudo-color) and 1H (grayscale) slices of right tumor receiving an intratumoral injection of 107 TATP-F68-PFC-labeled CAR T cells (A) and the left tumor with the same number of F68-PFC-labeled CAR T cells (B) |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
REFERENCES
- 1June CH, O'Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science. 2018; 359: 1361–1365.
- 2Townsend MH, Shrestha G, Robison RA, O'Neill KL. The expansion of targetable biomarkers for CAR T cell therapy. J Exp Clin Cancer Res. 2018; 37: 163.
- 3Bonifant CL, Jackson HJ, Brentjens RJ, Curran KJ. Toxicity and management in CAR T-cell therapy. Mol Ther Oncolytics. 2016; 3: 16011.
- 4Chang ZL, Chen YY. CARs: synthetic immunoreceptors for cancer therapy and beyond. Trends Mol Med. 2017; 23: 430–450.
- 5Hartmann J, Schüßler-Lenz M, Bondanza A, Buchholz CJ. Clinical development of CAR T cells—challenges and opportunities in translating innovative treatment concepts. EMBO Mol Med. 2017; 9:e201607485.
- 6Tarantal AF, Lee C, Kukis DL, Cherry SR. Radiolabeling human peripheral blood stem cells for positron emission tomography (PET) imaging in young rhesus monkeys. PLoS ONE. 2013; 8:e77148.
- 7Wolfs E, Struys T, Notelaers T, et al. 18F-FDG labeling of mesenchymal stem cells and multipotent adult progenitor cells for PET imaging: effects on ultrastructure and differentiation capacity. J Nucl Med. 2013; 54: 447–454.
- 8Wang X, Rosol M, Ge S, et al. Dynamic tracking of human hematopoietic stem cell engraftment using in vivo bioluminescence imaging. Blood. 2003; 102: 3478–3482.
- 9Xiong T, Zhang Z, Liu B-F, et al. In vivo optical imaging of human adenoid cystic carcinoma cell metastasis. Oral Oncol. 2005; 41: 709–715.
- 10Giepmans B, Adams SR, Ellisman MH, Tsien RY. The fluorescent toolbox for assessing protein location and function. Science. 2006; 312: 217–224.
- 11Ahrens ET, Bulte JW. Tracking immune cells in vivo using magnetic resonance imaging. Nat Rev Immunol. 2013; 13: 755–763.
- 12Ahrens ET, Helfer BM, O'Hanlon CF, Schirda C. Clinical cell therapy imaging using a perfluorocarbon tracer and fluorine-19 MRI. Magn Reson Med. 2014; 72: 1696–1701.
- 13Kircher MF, Gambhir SS, Grimm J. Noninvasive cell-tracking methods. Nat Rev Clin Oncol. 2011; 8: 677–688.
- 14Bouchlaka MN, Ludwig KD, Gordon JW, et al. (19)F-MRI for monitoring human NK cells in vivo. Oncoimmunology. 2016; 5:e1143996.
- 15Fink C, Gaudet JM, Fox MS, et al. (19)F-perfluorocarbon-labeled human peripheral blood mononuclear cells can be detected in vivo using clinical MRI parameters in a therapeutic cell setting. Sci Rep. 2018; 8: 590.
- 16Janjic JM, Ahrens ET. Fluorine-containing nanoemulsions for MRI cell tracking. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2009; 1: 492–501.
- 17Ku M-C, Edes I, Bendix I, et al. ERK1 as a therapeutic target for dendritic cell vaccination against high-grade gliomas. Mol Cancer Ther. 2016; 15: 1975–1987.
- 18Somanchi SS, Kennis BA, Gopalakrishnan V, Lee DA, Bankson JA. In vivo (19)F-magnetic resonance imaging of adoptively transferred NK cells. Methods Mol Biol. 2016; 1441: 317–332.
- 19Srinivas M, Turner MS, Janjic JM, Morel PA, Laidlaw DH, Ahrens ET. In vivo cytometry of antigen-specific t cells using 19F MRI. Magn Reson Med. 2009; 62: 747–753.
- 20Waiczies H, Lepore S, Janitzek N, et al. Perfluorocarbon particle size influences magnetic resonance signal and immunological properties of dendritic cells. PLoS ONE. 2011; 6:e21981.
- 21Xander Staal OK. Mangala Srinivas. Chapter 11: in vivo 19-fluorine magnetic resonance imaging. In: G Haufe, F Leroux, eds. Fluorine in Life Sciences: Pharmaceuticals, Medicinal Diagnostics, and Agrochemicals. 2019: 397–424.
10.1016/B978-0-12-812733-9.00011-8 Google Scholar
- 22Ruiz-Cabello J, Barnett BP, Bottomley PA, Bulte J. Fluorine (19F) MRS and MRI in biomedicine. NMR Biomed. 2011; 24: 114–129.
- 23Ahrens ET, Zhong J. In vivo MRI cell tracking using perfluorocarbon probes and fluorine-19 detection. NMR Biomed. 2013; 26: 860–871.
- 24Chapelin F, Capitini CM, Ahrens ET. Fluorine-19 MRI for detection and quantification of immune cell therapy for cancer. J Immunother Cancer. 2018; 6: 105.
- 25Lim WA, June CH. The principles of engineering immune cells to treat cancer. Cell. 2017; 168: 724–740.
- 26Lindgren M, Hällbrink M, Prochiantz A, Langel Ü. Cell-penetrating peptides. Trends Pharmacol Sci. 2000; 21: 99–103.
- 27Vives E, Brodin P, Lebleu B. A truncated HIV-1 Tat protein basic domain rapidly translocates through the plasma membrane and accumulates in the cell nucleus. J Biol Chem. 1997; 272: 16010–16017.
- 28Janjic JM, Srinivas M, Kadayakkara DK, Ahrens ET. Self-delivering nanoemulsions for dual fluorine-19 MRI and fluorescence detection. J Am Chem Soc. 2008; 130: 2832–2841.
- 29Ahrens ET, Flores R, Xu H, Morel PA. In vivo imaging platform for tracking immunotherapeutic cells. Nat Biotechnol. 2005; 23: 983–987.
- 30Johnson LA, Scholler J, Ohkuri T, et al. Rational development and characterization of humanized anti–EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci Transl Med. 2015; 7: 275ra222.
- 31Chapelin F, Gao S, Okada H, Weber TG, Messer K, Ahrens ET. Fluorine-19 nuclear magnetic resonance of chimeric antigen receptor T cell biodistribution in murine cancer model. Sci Rep. 2017; 7: 17748.
- 32Ohno M, Ohkuri T, Kosaka A, et al. Expression of miR-17-92 enhances anti-tumor activity of T-cells transduced with the anti-EGFRvIII chimeric antigen receptor in mice bearing human GBM xenografts. J Immunother Cancer. 2013; 1: 21.
- 33Srinivas M, Morel PA, Ernst LA, Laidlaw DH, Ahrens ET. Fluorine-19 MRI for visualization and quantification of cell migration in a diabetes model. Magn Reson Med. 2007; 58: 725–734.
- 34Howard MD, Hood ED, Greineder CF, Alferiev IS, Chorny M, Muzykantov V. Targeting to endothelial cells augments the protective effect of novel dual bioactive antioxidant/anti-inflammatory nanoparticles. Mol Pharm. 2014; 11: 2262–2270.
- 35Hu S, Wang T, Pei X, et al. Synergistic enhancement of antitumor efficacy by PEGylated Multi-walled carbon nanotubes modified with cell-penetrating peptide TAT. Nanoscale Res Lett. 2016; 11: 452.
- 36Hitchens TK, Liu L, Foley LM, Simplaceanu V, Ahrens ET, Ho C. Combining perfluorocarbon and superparamagnetic iron-oxide cell labeling for improved and expanded applications of cellular MRI. Magn Reson Med. 2015; 73: 367–375.
- 37van Eeden SF, Klut ME, Leal MA, Alexander J, Zonis Z, Skippen P. Partial liquid ventilation with perfluorocarbon in acute lung injury: light and transmission electron microscopy studies. Am J Respir Cell Mol Biol. 2000; 22: 441–450.
- 38Gonzales C, Yoshihara HAI, Dilek N, et al. In-vivo detection and tracking of T cells in various organs in a melanoma tumor model by 19F-fluorine MRS/MRI. PLoS ONE. 2016; 11:e0164557.
- 39Flogel U, Su S, Kreideweiss I, et al. Early detection of transplant rejection by in vivo 19F MRI. Circulation. 2009; 120: S817–S817.
- 40Ahrens ET, Young WB, Xu H, Pusateri LK. Rapid quantification of inflammation in tissue samples using perfluorocarbon emulsion and fluorine-19 nuclear magnetic resonance. Biotechniques. 2011; 50: 229–234.
- 41Khurana A, Chapelin F, Xu H, et al. Visualization of macrophage recruitment in head and neck carcinoma model using fluorine-19 magnetic resonance imaging. Magn Reson Med. 2018; 79: 1972–1980.
- 42Kadayakkara DK, Ranganathan S, Young WB, Ahrens ET. Assaying macrophage activity in a murine model of inflammatory bowel disease using fluorine-19 MRI. Lab Invest. 2012; 92: 636–645.
- 43Zhong J, Narsinh K, Morel PA, Xu H, Ahrens ET. In Vivo Quantification of inflammation in experimental autoimmune encephalomyelitis rats using Fluorine-19 magnetic resonance imaging reveals immune cell recruitment outside the nervous system. PLoS ONE. 2015; 10:e0140238.
- 44Temme S, Grapentin C, Quast C, et al. Noninvasive imaging of early venous thrombosis by 19F magnetic resonance imaging with targeted perfluorocarbon nanoemulsions. Circulation. 2015; 131: 1405–1414.
- 45Grapentin C, Mayenfels F, Barnert S, et al. Optimization of perfluorocarbon nanoemulsions for molecular imaging by 19F MRI. In: A Seifalin, A Mel, DM Kalaskar, eds. Nanomedicine. Manchester: One Central Press; 2014: 268–286.
- 46Temme S, Baran P, Bouvain P, et al. Synthetic cargo internalization receptor system for nanoparticle tracking of individual cell populations by fluorine magnetic resonance imaging. ACS Nano. 2018; 12: 11178–11192.
- 47Girotti AW. Mechanisms of lipid peroxidation. J Free Radic Biol Med. 1985; 1: 87–95.
- 48Schaich KM. Metals and lipid oxidation. Contemporary issues. Lipids. 1992; 27: 209–218.
- 49Fretz M, Jin J, Conibere R, et al. Effects of Na+/H+ exchanger inhibitors on subcellular localisation of endocytic organelles and intracellular dynamics of protein transduction domains HIV–TAT peptide and octaarginine. J Control Release. 2006; 116: 247–254.
- 50Patrick MJ, Janjic JM, Teng H, et al. Intracellular pH measurements using perfluorocarbon nanoemulsions. J Am Chem Soc. 2013; 135: 18445–18457.
- 51Castro O, Nesbitt AE, Lyles D. Effect of a perfluorocarbon emulsion (Fluosol-DA) on reticuloendothelial system clearance function. Am J Hematol. 1984; 16: 15–21.
- 52de la Fuente JM, Berry CC. Tat peptide as an efficient molecule to translocate gold nanoparticles into the cell nucleus. Bioconjug Chem. 2005; 16: 1176–1180.
- 53Krafft MP. Fluorocarbons and fluorinated amphiphiles in drug delivery and biomedical research. Adv Drug Deliv Rev. 2001; 47: 209–228.
- 54Krafft MP, Riess JG. Perfluorocarbons: life sciences and biomedical uses dedicated to the memory of Professor Guy Ourisson, a true RENAISSANCE man. J Polym Sci Pol Chem. 2007; 45: 1185–1198.
- 55Brooks H, Lebleu B, Vivès E. Tat peptide-mediated cellular delivery: back to basics. Adv Drug Deliv Rev. 2005; 57: 559–577.
- 56Mazel M, Clair P, Rousselle C, et al. Doxorubicin-peptide conjugates overcome multidrug resistance. Anticancer Drugs. 2001; 12: 107–116.
- 57Fawell S, Seery J, Daikh Y, et al. Tat-mediated delivery of heterologous proteins into cells. Proc Natl Acad Sci U S A. 1994; 91: 664–668.
- 58Torchilin VP, Levchenko TS, Rammohan R, Volodina N, Papahadjopoulos-Sternberg B, D'Souza G. Cell transfection in vitro and in vivo with nontoxic TAT peptide-liposome–DNA complexes. Proc Natl Acad Sci U S A. 2003; 100: 1972–1977.
- 59Foged C, Nielsen HM. Cell-penetrating peptides for drug delivery across membrane barriers. Expert Opin Drug Deliv. 2008; 5: 105–117.
- 60Boisguerin P, Redt-Clouet C, Franck-Miclo A, et al. Systemic delivery of BH4 anti-apoptotic peptide using CPPs prevents cardiac ischemia–reperfusion injuries in vivo. J Control Release. 2011; 156: 146–153.
- 61Borsello T, Clarke PGH, Hirt L, et al. A peptide inhibitor of c-Jun N-terminal kinase protects against excitotoxicity and cerebral ischemia. Nat Med. 2003; 9: 1180.
- 62Michiue H, Eguchi A, Scadeng M, Dowdy SF. Induction of in vivo synthetic lethal RNAi responses to treat glioblastoma. Cancer Biol Ther. 2009; 8: 2304–2311.
- 63Yang D, Sun Y-Y, Lin X, et al. Intranasal delivery of cell-penetrating anti-NF-κB peptides (Tat-NBD) alleviates infection-sensitized hypoxic–ischemic brain injury. Exp Neurol. 2013; 247: 447–455.
- 64Bates E, Bode C, et al. Direct Inhibition of delta-Protein Kinase C Enzyme to Limit Total Infarct Size in Acute Myocardial Infarction (DELTA MI) Investigators Intracoronary KAI-9803 as an adjunct to primary percutaneous coronary intervention for acute ST-segment elevation myocardial infarction. Circulation. 2008; 117: 886–896.
- 65Cousins MJ, Pickthorn K, Huang S, Critchley L, Bell G. The safety and efficacy of KAI-1678- an inhibitor of epsilon protein kinase C (epsilonPKC)-versus lidocaine and placebo for the treatment of postherpetic neuralgia: a crossover study design. Pain Med. 2013; 14: 533–540.
- 66Suckfuell M, Lisowska G, Domka W, et al. Efficacy and safety of AM-111 in the treatment of acute sensorineural hearing loss: a double-blind, randomized, placebo-controlled phase II study. Otol Neurotol. 2014; 35: 1317–1326.
- 67Deloche C, Lopez-Lazaro L, Mouz S, Perino J, Abadie C, Combette JM. XG-102 administered to healthy male volunteers as a single intravenous infusion: a randomized, double-blind, placebo-controlled, dose-escalating study. Pharmacol Res Perspect. 2014; 2:e00020.
- 68Tatsumi T, Huang J, Gooding WE, et al. Intratumoral delivery of dendritic cells engineered to secrete both interleukin (IL)-12 and IL-18 effectively treats local and distant disease in association with broadly reactive Tcl-type immunity. Cancer Res. 2003; 63: 6378–6386.
- 69Ngwa W, Irabor OC, Schoenfeld JD, Hesser J, Demaria S, Formenti SC. Using immunotherapy to boost the abscopal effect. Nat Rev Cancer. 2018; 18: 313–322.