Loading of CAR-T cells with magnetic nanoparticles for controlled targeting suppresses inflammatory cytokine release and switches tumor cell death mechanism

Felix Pfister , Lucas R. Carnell , Lisa Löffler , Philipp Boosz , Niels Schaft , Jan Dörrie , René Stein , Malte Lenz , Erdmann Spiecker , Christian M. Huber , Sami Haddadin , Carola Berking , Christoph Alexiou , Christina Janko

MedComm ›› 2025, Vol. 6 ›› Issue (1) : e70039

PDF
MedComm ›› 2025, Vol. 6 ›› Issue (1) : e70039 DOI: 10.1002/mco2.70039
ORIGINAL ARTICLE

Loading of CAR-T cells with magnetic nanoparticles for controlled targeting suppresses inflammatory cytokine release and switches tumor cell death mechanism

Author information +
History +
PDF

Abstract

Therapies against hematological malignancies using chimeric antigen receptors (CAR)-T cells have shown great potential; however, therapeutic success in solid tumors has been constrained due to limited tumor trafficking and infiltration, as well as the scarcity of cancer-specific solid tumor antigens. Therefore, the enrichment of tumor-antigen specific CAR-T cells in the desired region is critical for improving therapy efficacy and reducing systemic on-target/off-tumor side effects. Here, we functionalized human CAR-T cells with superparamagnetic iron oxide nanoparticles (SPIONs), making them magnetically controllable for site-directed targeting. SPION-loaded CAR-T cells maintained their specific cytolytic capacity against melanoma cells expressing the CAR-specific antigen chondroitin sulfate proteoglycan (CSPG4). Importantly, SPIONs suppressed cytokine release in the loaded CAR-T cells, shifting the cell death phenotype in the tumor cells from pyroptosis to apoptosis. Furthermore, SPION-loaded CAR-T cells could be enriched in a dynamic flow model through an external magnetic field and be detected in MRI. These results demonstrate that lytic cytotoxicity is retained after SPION-functionalization and provides a basis for future site-specific immunotherapies against solid tumors with reduced systemic adverse side effects.

Keywords

adoptive T cell therapy / cancer / CAR-T cell / magnetic cell targeting / pyroptosis / superparamagnetic iron oxide nanoparticles (SPIONs)

Cite this article

Download citation ▾
Felix Pfister, Lucas R. Carnell, Lisa Löffler, Philipp Boosz, Niels Schaft, Jan Dörrie, René Stein, Malte Lenz, Erdmann Spiecker, Christian M. Huber, Sami Haddadin, Carola Berking, Christoph Alexiou, Christina Janko. Loading of CAR-T cells with magnetic nanoparticles for controlled targeting suppresses inflammatory cytokine release and switches tumor cell death mechanism. MedComm, 2025, 6(1): e70039 DOI:10.1002/mco2.70039

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Bray F, Laversanne M, Sung H, et al. Global cancer statistics 2022: gLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024; 74(3): 229-263.

[2]

Hosseinkhani N, Derakhshani A, Kooshkaki O, et al. Immune checkpoints and CAR-T cells: the pioneers in future cancer therapies?. Int J Mol Sci. 2020; 21(21): 8305.

[3]

Levine BL. Performance-enhancing drugs: design and production of redirected chimeric antigen receptor (CAR) T cells. Cancer Gene Ther. 2015; 22(2): 79-84.

[4]

Levine BL, Miskin J, Wonnacott K, Keir C. Global manufacturing of CAR T cell therapy. Mol Ther Methods Clin Dev. 2017; 4: 92-101.

[5]

Martin T, Usmani SZ, Berdeja JG, et al. Ciltacabtagene autoleucel, an anti-B-cell maturation antigen chimeric antigen receptor T-cell therapy, for relapsed/refractory multiple myeloma: cARTITUDE-1 2-year follow-up. J Clin Oncol. 2023; 41(6): 1265-1274.

[6]

Chen YJ, Abila B, Kamel YM. CAR-T: what is next?. Cancers (Basel). 2023; 15(3): 663.

[7]

Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 2021; 11(4): 69.

[8]

Sadelain M, Brentjens R, Riviere I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013; 3(4): 388-398.

[9]

Curran KJ, Pegram HJ, Brentjens RJ. Chimeric antigen receptors for T cell immunotherapy: current understanding and future directions. J Gene Med. 2012; 14(6): 405-415.

[10]

Bonifant CL, Jackson HJ, Brentjens RJ, Curran KJ. Toxicity and management in CAR T-cell therapy. Mol Ther Oncolytics. 2016; 3: 16011.

[11]

Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood. 2016; 127(26): 3321-3330.

[12]

Hou AJ, Chen LC, Chen YY. Navigating CAR-T cells through the solid-tumour microenvironment. Nat Rev Drug Discov. 2021; 20(7): 531-550.

[13]

Kloss CC, Condomines M, Cartellieri M, Bachmann M, Sadelain M. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat Biotechnol. 2013; 31(1): 71-75.

[14]

Zhao Y, Moon E, Carpenito C, et al. Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor. Cancer Res. 2010; 70(22): 9053-9061.

[15]

Brunke O, Odenbach S, Jurgons R, Alexiou C, Hilger I, Beckmann F. Determination of the magnetic particle distribution in tumour tissue by means of x-ray tomography. J Phys: Condens Matter. 2006; 18: S2903.

[16]

Tietze R, Rahn H, Lyer S, et al. Visualization of superparamagnetic nanoparticles in vascular tissue using XmuCT and histology. Histochem Cell Biol. 2011; 135(2): 153-158.

[17]

Unterweger H, Janko C, Schwarz M, et al. Non-immunogenic dextran-coated superparamagnetic iron oxide nanoparticles: a biocompatible, size-tunable contrast agent for magnetic resonance imaging. Int J Nanomedicine. 2017; 12: 5223-5238.

[18]

Jin H, Qian Y, Dai Y, et al. Magnetic enrichment of dendritic cell vaccine in lymph node with fluorescent-magnetic nanoparticles enhanced cancer immunotherapy. Theranostics. 2016; 6(11): 2000-2014.

[19]

Polyak B, Fishbein I, Chorny M, et al. High field gradient targeting of magnetic nanoparticle-loaded endothelial cells to the surfaces of steel stents. Proc Natl Acad Sci USA. 2008; 105(2): 698-703.

[20]

Tukmachev D, Lunov O, Zablotskii V, et al. An effective strategy of magnetic stem cell delivery for spinal cord injury therapy. Nanoscale. 2015; 7(9): 3954-3958.

[21]

Kiru L, Zlitni A, Tousley AM, et al. In vivo imaging of nanoparticle-labeled CAR T cells. Proc Natl Acad Sci USA. 2022; 119(6): e2102363119.

[22]

Boosz P, Pfister F, Stein R, et al. Citrate-coated superparamagnetic iron oxide nanoparticles enable a stable non-spilling loading of T cells and their magnetic accumulation. Cancers (Basel). 2021; 13(16): 4143.

[23]

Mühlberger M, Janko C, Unterweger H, et al. Functionalization of T lymphocytes with citrate-coated superparamagnetic iron oxide nanoparticles for magnetically controlled immune therapy. Int J Nanomedicine. 2019; 14: 8421-8432.

[24]

Mühlberger M, Janko C, Unterweger H, et al. Functionalization of T lymphocytes for magnetically controlled immune therapy: selection of suitable superparamagnetic iron oxide nanoparticles. J Magnet Magnetic Mater. 2019; 473: 61-67.

[25]

Mühlberger M, Unterweger H, Band J, et al. Loading of primary human T lymphocytes with citrate-coated superparamagnetic iron oxide nanoparticles does not impair their activation after polyclonal stimulation. Cells. 2020; 9(2): 342.

[26]

Nie W, Wei W, Zuo L, et al. Magnetic nanoclusters armed with responsive PD-1 antibody synergistically improved adoptive T-cell therapy for solid tumors. ACS Nano. 2019; 13(2): 1469-1478.

[27]

Pai A, Cao P, White EE, et al. Dynamically programmable magnetic fields for controlled movement of cells loaded with iron oxide nanoparticles. ACS Appl Bio Mater. 2020; 3(7): 4139-4147.

[28]

Sanz-Ortega L, Portilla Y, Pérez-Yagüe S, Barber DF. Magnetic targeting of adoptively transferred tumour-specific nanoparticle-loaded CD8(+) T cells does not improve their tumour infiltration in a mouse model of cancer but promotes the retention of these cells in tumour-draining lymph nodes. J Nanobiotechnol. 2019; 17(1): 87.

[29]

Sanz-Ortega L, Rojas JM, Marcos A, Portilla Y, Stein JV, Barber DF. T cells loaded with magnetic nanoparticles are retained in peripheral lymph nodes by the application of a magnetic field. J Nanobiotechnol. 2019; 17(1): 14.

[30]

Sanz-Ortega L, Rojas JM, Portilla Y, Perez-Yague S, Barber DF. Magnetic nanoparticles attached to the NK cell surface for tumor targeting in adoptive transfer therapies does not affect cellular effector functions. Front Immunol. 2019; 10: 2073.

[31]

Pfister F, Dorrie J, Schaft N, et al. Human T cells loaded with superparamagnetic iron oxide nanoparticles retain antigen-specific TCR functionality. Front Immunol. 2023; 14: 1223695.

[32]

Behm FG, Smith FO, Raimondi SC, Pui CH, Bernstein ID. Human homologue of the rat chondroitin sulfate proteoglycan, NG2, detected by monoclonal antibody 7.1, identifies childhood acute lymphoblastic leukemias with t(4;11)(q21;q23) or t(11;19)(q23;p13) and MLL gene rearrangements. Blood. 1996; 87(3): 1134-1139.

[33]

Chekenya M, Rooprai HK, Davies D, Levine JM, Butt AM, Pilkington GJ. The NG2 chondroitin sulfate proteoglycan: role in malignant progression of human brain tumours. Int J Dev Neurosci. 1999; 17(5-6): 421-435.

[34]

Godal A, Bruland O, Haug E, Aas M, Fodstad O. Unexpected expression of the 250 kD melanoma-associated antigen in human sarcoma cells. Br J Cancer. 1986; 53(6): 839-841.

[35]

Shoshan Y, Nishiyama A, Chang A, et al. Expression of oligodendrocyte progenitor cell antigens by gliomas: implications for the histogenesis of brain tumors. Proc Natl Acad Sci USA. 1999; 96(18): 10361-10366.

[36]

Challier JC, Carbillon L, Kacemi A, et al. Characterization of first trimester human fetal placental vessels using immunocytochemical markers. Cell Mol Biol (Noisy-le-grand). 2001; 47. Online Pub: OL79-87.

[37]

Midwood KS, Salter DM. Expression of NG2/human melanoma proteoglycan in human adult articular chondrocytes. Osteoarthritis Cartilage. 1998; 6(5): 297-305.

[38]

Petrini S, Tessa A, Carrozzo R, et al. Human melanoma/NG2 chondroitin sulfate proteoglycan is expressed in the sarcolemma of postnatal human skeletal myofibers. Abnormal expression in merosin-negative and Duchenne muscular dystrophies. Mol Cell Neurosci. 2003; 23(2): 219-231.

[39]

Tordsson JM, Ohlsson LG, Abrahmsen LB, Karlstrom PJ, Lando PA, Brodin TN. Phage-selected primate antibodies fused to superantigens for immunotherapy of malignant melanoma. Cancer Immunol Immunother. 2000; 48(12): 691-702.

[40]

Ferrone S, Chen ZJ, Liu CC, Hirai S, Kageshita T, Mittelman A. Human high molecular weight-melanoma associated antigen mimicry by mouse anti-idiotypic monoclonal antibodies MK2-23. Experimental studies and clinical trials in patients with malignant melanoma. Pharmacol Ther. 1993; 57(2-3): 259-290.

[41]

Schlingemann RO, Rietveld FJ, de Waal RM, Ferrone S, Ruiter DJ. Expression of the high molecular weight melanoma-associated antigen by pericytes during angiogenesis in tumors and in healing wounds. Am J Pathol. 1990; 136(6): 1393-1405.

[42]

Krug C, Birkholz K, Paulus A, et al. Stability and activity of MCSP-specific chimeric antigen receptors (CARs) depend on the scFv antigen-binding domain and the protein backbone. Cancer Immunol Immunother. 2015; 64(12): 1623-1635.

[43]

Charpentier JC, King PD. Mechanisms and functions of endocytosis in T cells. Cell Commun Signal. 2021; 19(1): 92.

[44]

Rodriguez-Marquez P, Calleja-Cervantes ME, Serrano G, et al. CAR density influences antitumoral efficacy of BCMA CAR T cells and correlates with clinical outcome. Sci Adv. 2022; 8(39): eabo0514.

[45]

Ho JY, Wang L, Liu Y, et al. Promoter usage regulating the surface density of CAR molecules may modulate the kinetics of CAR-T cells in vivo. Mol Ther Methods Clin Dev. 2021; 21: 237-246.

[46]

Krug C, Wiesinger M, Abken H, et al. A GMP-compliant protocol to expand and transfect cancer patient T cells with mRNA encoding a tumor-specific chimeric antigen receptor. Cancer Immunol Immunother. 2014; 63(10): 999-1008.

[47]

Morris EC, Neelapu SS, Giavridis T, Sadelain M. Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nat Rev Immunol. 2022; 22(2): 85-96.

[48]

Wiesinger M, Marz J, Kummer M, et al. Clinical-scale production of CAR-T cells for the treatment of melanoma patients by mRNA transfection of a CSPG4-specific CAR under full GMP compliance. Cancers (Basel). 2019; 11(8): 1198.

[49]

Dorrie J, Babalija L, Hoyer S, et al. BRAF and MEK inhibitors influence the function of reprogrammed T cells: consequences for adoptive T-cell therapy. Int J Mol Sci. 2018; 19(1): 289.

[50]

Miyawaki T, Kasahara Y, Kanegane H, et al. Expression of CD45R0 (UCHL1) by CD4+ and CD8+ T cells as a sign of in vivo activation in infectious mononucleosis. Clin Exp Immunol. 1991; 83(3): 447-451.

[51]

Deng T, Tang C, Zhang G, Wan X. DAMPs released by pyroptotic cells as major contributors and therapeutic targets for CAR-T-related toxicities. Cell Death Dis. 2021; 12(1): 129.

[52]

Liu Y, Fang Y, Chen X, et al. Gasdermin E-mediated target cell pyroptosis by CAR T cells triggers cytokine release syndrome. Sci Immunol. 2020; 5(43): eaax7969.

[53]

Yang H, Wang H, Wen C, et al. Effects of iron oxide nanoparticles as T(2)-MRI contrast agents on reproductive system in male mice. J Nanobiotechnol. 2022; 20(1): 98.

[54]

Bulte JWM, Daldrup-Link HE. Clinical tracking of cell transfer and cell transplantation: trials and tribulations. Radiology. 2018; 289(3): 604-615.

[55]

Bruni D, Angell HK, Galon J. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat Rev Cancer. 2020; 20(11): 662-680.

[56]

Waiczies S, Niendorf T, Lombardi G. Labeling of cell therapies: how can we get it right?. Oncoimmunology. 2017; 6(10): e1345403.

[57]

Panet E, Mashriki T, Lahmi R, et al. The interface of nanoparticles with proliferating mammalian cells. Nat Nanotechnol. 2017; 12(7): 598-600.

[58]

McMahon HT, Boucrot E. Molecular mechanism and physiological functions of clathrin-mediated endocytosis. Nat Rev Mol Cell Biol. 2011; 12(8): 517-533.

[59]

Berger C, Rausch M, Schmidt P, Rudin M. Feasibility and limits of magnetically labeling primary cultured rat T cells with ferumoxides coupled with commonly used transfection agents. Mol Imaging. 2006; 5(2): 93-104.

[60]

Montet-Abou K, Montet X, Weissleder R, Josephson L. Transfection agent induced nanoparticle cell loading. Mol Imaging. 2005; 4(3): 165-171.

[61]

West DL, White SB, Zhang Z, Larson AC, Omary RA. Assessment and optimization of electroporation-assisted tumoral nanoparticle uptake in a nude mouse model of pancreatic ductal adenocarcinoma. Int J Nanomedicine. 2014; 9: 4169-4176.

[62]

Cieri N, Camisa B, Cocchiarella F, et al. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood. 2013; 121(4): 573-584.

[63]

Salter AI, Rajan A, Kennedy JJ, et al. Comparative analysis of TCR and CAR signaling informs CAR designs with superior antigen sensitivity and in vivo function. Sci Signal. 2021; 14(697): eabe2606.

[64]

Nerreter T, Letschert S, Gotz R, et al. Super-resolution microscopy reveals ultra-low CD19 expression on myeloma cells that triggers elimination by CD19 CAR-T. Nat Commun. 2019; 10(1): 3137.

[65]

Watanabe K, Terakura S, Martens AC, et al. Target antigen density governs the efficacy of anti-CD20-CD28-CD3 zeta chimeric antigen receptor-modified effector CD8+ T cells. J Immunol. 2015; 194(3): 911-920.

[66]

Zhai Y, Du Y, Li G, et al. Trogocytosis of CAR molecule regulates CAR-T cell dysfunction and tumor antigen escape. Signal Transduct Target Ther. 2023; 8(1): 457.

[67]

Liadi I, Singh H, Romain G, et al. Individual motile CD4(+) T cells can participate in efficient multikilling through conjugation to multiple tumor cells. Cancer Immunol Res. 2015; 3(5): 473-482.

[68]

Sommermeyer D, Hudecek M, Kosasih PL, et al. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia. 2016; 30(2): 492-500.

[69]

Shi J, Zhao Y, Wang K, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015; 526(7575): 660-665.

[70]

Chen X, He WT, Hu L, et al. Pyroptosis is driven by non-selective gasdermin-D pore and its morphology is different from MLKL channel-mediated necroptosis. Cell Res. 2016; 26(9): 1007-1020.

[71]

Zhang Z, Zhang Y, Xia S, et al. Gasdermin E suppresses tumour growth by activating anti-tumour immunity. Nature. 2020; 579(7799): 415-420.

[72]

Zhou Z, He H, Wang K, et al. Granzyme A from cytotoxic lymphocytes cleaves GSDMB to trigger pyroptosis in target cells. Science. 2020; 368(6494).

[73]

Xiao X, Huang S, Chen S, et al. Mechanisms of cytokine release syndrome and neurotoxicity of CAR T-cell therapy and associated prevention and management strategies. J Exp Clin Cancer Res. 2021; 40(1): 367.

[74]

Wang J-L, Hua S-N, Bao H-J, Yuan J, Zhao Y, Chen S. Pyroptosis and inflammasomes in cancer and inflammation. MedComm. 2023; 4(5): e374.

[75]

Tietze R, Lyer S, Durr S, et al. Efficient drug-delivery using magnetic nanoparticles–biodistribution and therapeutic effects in tumour bearing rabbits. Nanomedicine. 2013; 9(7): 961-971.

[76]

Harrer DC, Dorrie J, Schaft N. CSPG4 as target for CAR-T-cell therapy of various tumor entities-merits and challenges. Int J Mol Sci. 2019; 20(23): 5942.

[77]

Gerer KF, Hoyer S, Dorrie J, Schaft N. Electroporation of mRNA as universal technology platform to transfect a variety of primary cells with antigens and functional proteins. Methods Mol Biol. 2017; 1499: 165-178.

[78]

Fedorov A, Beichel R, Kalpathy-Cramer J, et al. 3D Slicer as an image computing platform for the quantitative imaging network. Magn Reson Imaging. 2012; 30(9): 1323-1341.

[79]

Kappes M, Friedrich B, Pfister F, et al. Superparamagnetic iron oxide nanoparticles for targeted cell seeding: magnetic patterning and magnetic 3D cell culture. Adv Funct Mater. 2022; 32(50): 2203672.

[80]

Behr J, Carnell LR, Stein R, et al. In vitro setup for determination of nanoparticle-mediated magnetic cell and drug accumulation in tumor spheroids under flow conditions. Cancers (Basel). 2022; 14(23): 5978.

[81]

Sauvola J, Pietikäinen M. Adaptive document image binarization. Pattern Recogn. 2000; 33(2): 225-236.

RIGHTS & PERMISSIONS

2025 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

201

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/