Leveraging biomaterials for enhancing T cell immunotherapy
Tài liệu tham khảo
Weber, 2020, The emerging landscape of immune cell therapies, Cell, 181, 46, 10.1016/j.cell.2020.03.001
O'Donnell, 2019, Cancer immunoediting and resistance to T cell-based immunotherapy, Nat. Rev. Clin. Oncol., 16, 151, 10.1038/s41571-018-0142-8
Couzin-Frankel, 2013, Breakthrough of the year 2013, Cancer Immunother. Sci., 342, 1432
Wraith, 2017, The future of immunotherapy: a 20-year perspective, Front. Immunol., 8, 1668, 10.3389/fimmu.2017.01668
Kumar, 2018, Human T cell development, localization, and function throughout life, Immunity, 48, 202, 10.1016/j.immuni.2018.01.007
Malissen, 2015, Early T cell activation: integrating biochemical, structural, and biophysical cues, Annu. Rev. Immunol., 33, 539, 10.1146/annurev-immunol-032414-112158
Masopust, 2013, The integration of T cell migration, differentiation and function, Nat. Rev. Immunol., 13, 309, 10.1038/nri3442
Zhang, 2011, CD8+ T cells: foot soldiers of the immune system, Immunity, 35, 161, 10.1016/j.immuni.2011.07.010
Takaba, 2017, The mechanisms of T cell selection in the thymus, Trends Immunol., 38, 805, 10.1016/j.it.2017.07.010
Kurd, 2016, T cell selection in the thymus: a spatial and temporal perspective, Immunol. Rev., 271, 114, 10.1111/imr.12398
Sakaguchi, 2008, Regulatory T cells and immune tolerance, Cell, 133, 775, 10.1016/j.cell.2008.05.009
Jiang, 2006, Regulation of immune responses by T cells, N. Engl. J. Med., 354, 1166, 10.1056/NEJMra055446
Sakaguchi, 2010, FOXP3 + regulatory T cells in the human immune system, Nat. Rev. Immunol., 10, 490, 10.1038/nri2785
Seder, 1994, Acquisition of lymphokine-producing phenotype by CD4+ T cells, Annu. Rev. Immunol., 12, 635, 10.1146/annurev.iy.12.040194.003223
Appay, 2002, Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections, Nat. Med., 8, 379, 10.1038/nm0402-379
Sahin, 2018, Personalized vaccines for cancer immunotherapy, Science, 359, 1355, 10.1126/science.aar7112
Buchbinder, 2016, CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition, Am. J. Clin. Oncol., 39, 98, 10.1097/COC.0000000000000239
Bluestone, 2005, Regulatory T-cell therapy: is it ready for the clinic?, Nat. Rev. Immunol., 5, 343, 10.1038/nri1574
Silva-Santos, 2015, γδ T cells in cancer, Nat. Rev. Immunol., 15, 683, 10.1038/nri3904
Gong, 2021, Nanomaterials for T-cell cancer immunotherapy, Nat. Nanotechnol., 16, 25, 10.1038/s41565-020-00822-y
Gaissmaier, 2020, Breaking bottlenecks for the TCR therapy of cancer, Cells, 9, 2095, 10.3390/cells9092095
Newick, 2017, CAR T cell therapy for solid tumors, Annu. Rev. Med., 68, 139, 10.1146/annurev-med-062315-120245
Wang, 2017, New development in CAR-T cell therapy, J. Hematol. Oncol., 10, 53, 10.1186/s13045-017-0423-1
Hinrichs, 2014, Exploiting the curative potential of adoptive T-cell therapy for cancer, Immunol. Rev., 257, 56, 10.1111/imr.12132
June, 2007, Adoptive T cell therapy for cancer in the clinic, J. Clin. Invest., 117, 1466, 10.1172/JCI32446
Leen, 2007, Improving T cell therapy for cancer, Annu. Rev. Immunol., 25, 243, 10.1146/annurev.immunol.25.022106.141527
Ferreira, 2019, Next-generation regulatory T cell therapy, Nat. Rev. Drug Discov., 18, 749, 10.1038/s41573-019-0041-4
Zhao, 2019, Engineered T cell therapy for cancer in the clinic, Front. Immunol., 10, 2250, 10.3389/fimmu.2019.02250
Gee, 2018, Antigen identification for orphan t cell receptors expressed on tumor-infiltrating lymphocytes, Cell, 172, 549, 10.1016/j.cell.2017.11.043
Huang, 2020, Recent advances in CAR-T cell engineering, J. Hematol. Oncol., 13, 86, 10.1186/s13045-020-00910-5
Biernacki, 2019, T-cell receptor-based immunotherapy for hematologic malignancies, Cancer J., 25, 179, 10.1097/PPO.0000000000000378
Ni, 2021, Clinical and basic research progress on Treg-induced immune tolerance in liver transplantation, Front. Immunol., 12, 535012, 10.3389/fimmu.2021.535012
June, 2015, Adoptive cellular therapy: a race to the finish line, Sci. Transl. Med., 7, 10.1126/scitranslmed.aaa3643
Shah, 2019, Mechanisms of resistance to CAR T cell therapy, Nat. Rev. Clin. Oncol., 16, 372
Bonifant, 2016, Toxicity and management in CAR T-cell therapy, Mol. Ther., 3, 16011
Levine, 2017, Global manufacturing of CAR T cell therapy, Mol. Ther., 4, 92
Hutmacher, 2010, Biomaterials offer cancer research the third dimension, Nat. Mater., 9, 90, 10.1038/nmat2619
Pashuck, 2012, Designing regenerative biomaterial therapies for the clinic, Sci. Transl. Med., 4, 10.1126/scitranslmed.3002717
Zhang, 2018, Nanomedicine for obesity treatment, Sci. China, 61, 373, 10.1007/s11427-017-9257-1
Jaganathan, 2014, Biomaterials in cardiovascular research: applications and clinical implications, BioMed Res. Int., 2014, 459465, 10.1155/2014/459465
Cheng, 2010, Nanoparticulate cellular patches for cell-mediated tumoritropic delivery, ACS Nano, 4, 625, 10.1021/nn901319y
Shen, 2021, A nanotherapeutic strategy to overcome chemotherapeutic resistance of cancer stem-like cells, Nat. Nanotechnol., 16, 104, 10.1038/s41565-020-00793-0
Fenton, 2018, Advances in biomaterials for drug delivery, Adv. Mater., 30, 10.1002/adma.201705328
Lu, 2016, Bioresponsive materials, Nat. Rev. Mater., 2, 10.1038/natrevmats.2016.75
Wang, 2017, Tailoring biomaterials for cancer immunotherapy: emerging trends and future outlook, Adv. Mater., 29
Wang, 2018, Biomaterial-assisted targeted modulation of immune cells in cancer treatment, Nat. Mater., 17, 761, 10.1038/s41563-018-0147-9
Dellacherie, 2019, Macroscale biomaterials strategies for local immunomodulation, Nat. Rev. Mater., 4, 379, 10.1038/s41578-019-0106-3
Lokugamage, 2019, Constrained nanoparticles deliver siRNA and sgRNA to T cells in vivo without targeting ligands, Adv. Mater., 31, 10.1002/adma.201902251
Tibbitt, 2009, Hydrogels as extracellular matrix mimics for 3D cell culture, Biotechnol. Bioeng., 103, 655, 10.1002/bit.22361
Isser, 2021, Biomaterials to enhance antigen-specific T cell expansion for cancer immunotherapy, Biomaterials, 268, 10.1016/j.biomaterials.2020.120584
Lee, 2015, T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial, Lancet, 385, 517, 10.1016/S0140-6736(14)61403-3
Rosenberg, 2008, Adoptive cell transfer: a clinical path to effective cancer immunotherapy, Nat. Rev. Cancer, 8, 299, 10.1038/nrc2355
Khalil, 2015, The new era of cancer immunotherapy: manipulating T-cell activity to overcome malignancy, Adv. Cancer Res., 128, 1, 10.1016/bs.acr.2015.04.010
Eggermont, 2014, Towards efficient cancer immunotherapy: advances in developing artificial antigen-presenting cells, Trends Biotechnol., 32, 456, 10.1016/j.tibtech.2014.06.007
Smith-Garvin, 2009, T cell activation, Annu. Rev. Immunol., 27, 591, 10.1146/annurev.immunol.021908.132706
Ross, 2018, Signaling and function of interleukin-2 in T lymphocytes, Annu. Rev. Immunol., 36, 411, 10.1146/annurev-immunol-042617-053352
Wolfl, 2014, Antigen-specific activation and cytokine-facilitated expansion of naive, human CD8+ T cells, Nat. Protoc., 9, 950, 10.1038/nprot.2014.064
Mackensen, 2006, Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T cells for the treatment of patients with metastatic melanoma, J. Clin. Oncol., 24, 5060, 10.1200/JCO.2006.07.1100
Hunder, 2008, Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1, N. Engl. J. Med., 358, 2698, 10.1056/NEJMoa0800251
Cheung, 2018, Scaffolds that mimic antigen-presenting cells enable ex vivo expansion of primary T cells, Nat. Biotechnol., 36, 160, 10.1038/nbt.4047
Butler, 2014, Human cell-based artificial antigen-presenting cells for cancer immunotherapy, Immunol. Rev., 257, 191, 10.1111/imr.12129
Zhang, 2007, 4-1BB is superior to CD28 costimulation for generating CD8+ cytotoxic lymphocytes for adoptive immunotherapy, J. Immunol., 179, 4910, 10.4049/jimmunol.179.7.4910
Gong, 2008, Establishment and characterization of a cell based artificial antigen-presenting cell for expansion and activation of CD8+ T cells ex vivo, Cell. Mol. Immunol., 5, 47, 10.1038/cmi.2008.6
Maus, 2002, Ex vivo expansion of polyclonal and antigen-specific cytotoxic T lymphocytes by artificial APCs expressing ligands for the T-cell receptor, CD28 and 4-1BB, Nat. Biotechnol., 20, 143, 10.1038/nbt0202-143
Hippen, 2008, Umbilical cord blood regulatory T-cell expansion and functional effects of tumor necrosis factor receptor family members OX40 and 4-1BB expressed on artificial antigen-presenting cells, Blood, 112, 2847, 10.1182/blood-2008-01-132951
Schmidts, 2020, Cell-based artificial APC resistant to lentiviral transduction for efficient generation of CAR-T cells from various cell sources, J. Immunother. Cancer, 8, 10.1136/jitc-2020-000990
Wang, 2017, Red blood cells for glucose-responsive insulin delivery, Adv. Mater., 29
Sun, 2020, DNA-edited ligand positioning on red blood cells to enable optimized T cell activation for adoptive immunotherapy, Angew. Chem. Int. Ed. Engl., 59, 14842, 10.1002/anie.202003367
Kim, 2004, The ABCs of artificial antigen presentation, Nat. Biotechnol., 22, 403, 10.1038/nbt955
Cai, 1996, Transfected drosophila cells as a probe for defining the minimal requirements for stimulating unprimed CD8+ T cells, Proc. Natl. Acad. Sci. U. S. A., 93, 14736, 10.1073/pnas.93.25.14736
Latouche, 2000, Induction of human cytotoxic T lymphocytes by artificial antigen-presenting cells, Nat. Biotechnol., 18, 405, 10.1038/74455
Dunn, 2019, T cell immunotherapy enhanced by designer biomaterials, Biomaterials, 217, 119265, 10.1016/j.biomaterials.2019.119265
Trickett, 2003, T cell stimulation and expansion using anti-CD3/CD28 beads, J. Immunol. Methods, 275, 251, 10.1016/S0022-1759(03)00010-3
Kalamasz, 2004, Optimization of human T-cell expansion ex vivo using magnetic beads conjugated with anti-CD3 and Anti-CD28 antibodies, J. Immunother., 27, 405, 10.1097/00002371-200409000-00010
Neurauter, 2007, Cell isolation and expansion using Dynabeads, Adv. Biochem. Eng. Biotechnol., 106, 41
Oelke, 2003, Ex vivo induction and expansion of antigen-specific cytotoxic T cells by HLA-Ig-coated artificial antigen-presenting cells, Nat. Med., 9, 619, 10.1038/nm869
Caserta, 2008, Synthetic CD4+ T cell-targeted antigen-presenting cells elicit protective antitumor responses, Cancer. Res., 68, 3010, 10.1158/0008-5472.CAN-07-5796
Clemente-Casares, 2016, Expanding antigen-specific regulatory networks to treat autoimmunity, Nature, 530, 434, 10.1038/nature16962
Singha, 2017, Peptide-MHC-based nanomedicines for autoimmunity function as T-cell receptor microclustering devices, Nat. Nanotechnol., 12, 701, 10.1038/nnano.2017.56
Vogt, 2002, Clustering of MHC-peptide complexes prior to their engagement in the immunological synapse: lipid raft and tetraspan microdomains, Immunol. Rev., 189, 136, 10.1034/j.1600-065X.2002.18912.x
Mossman, 2005, Altered TCR signaling from geometrically repatterned immunological synapses, Science, 310, 1191, 10.1126/science.1119238
Basu, 2017, Mechanical communication at the immunological synapse, Trends Cell Biol., 27, 241, 10.1016/j.tcb.2016.10.005
Doh, 2006, Immunological synapse arrays: patterned protein surfaces that modulate immunological synapse structure formation in T cells, Proc. Natl. Acad. Sci. U. S. A., 103, 5700, 10.1073/pnas.0509404103
Zhang, 2017, Biomimetic magnetosomes as versatile artificial antigen-presenting cells to potentiate T-cell-based anticancer therapy, ACS Nano, 11, 10724, 10.1021/acsnano.7b04955
Cheng, 2020, Artificial mini dendritic cells boost T cell-based immunotherapy for ovarian cancer, Adv. Sci. (Weinheim, Ger.), 7
Hickey, 2018, Efficient magnetic enrichment of antigen-specific T cells by engineering particle properties, Biomaterials, 187, 105, 10.1016/j.biomaterials.2018.09.029
Hickey, 2017, Biologically inspired design of nanoparticle artificial antigen-presenting cells for immunomodulation, Nano Lett., 17, 7045, 10.1021/acs.nanolett.7b03734
Matic, 2013, Fine tuning and efficient T cell activation with stimulatory aCD3 nanoarrays, Nano Lett., 13, 5090, 10.1021/nl4022623
Hammink, 2021, Semiflexible immunobrushes induce enhanced T cell activation and expansion, ACS Appl. Mater. Interfaces, 13, 16007, 10.1021/acsami.0c21994
Anderson, 2000, Concentration of MHC class II molecules in lipid rafts facilitates antigen presentation, Nat. Immunol., 1, 156, 10.1038/77842
Zappasodi, 2008, The effect of artificial antigen-presenting cells with preclustered anti-CD28/-CD3/-LFA-1 monoclonal antibodies on the induction of ex vivo expansion of functional human antitumor T cells, Haematologica, 93, 1523, 10.3324/haematol.12521
Perica, 2014, Magnetic field-induced T cell receptor clustering by nanoparticles enhances T cell activation and stimulates antitumor activity, ACS Nano, 8, 2252, 10.1021/nn405520d
Kosmides, 2018, Separating T cell targeting components onto magnetically clustered nanoparticles boosts activation, Nano Lett., 18, 1916, 10.1021/acs.nanolett.7b05284
Majedi, 2019, Augmentation of T-cell activation by oscillatory forces and engineered antigen-presenting cells, Nano Lett., 19, 6945, 10.1021/acs.nanolett.9b02252
Yang, 2020, Size-transformable antigen-presenting cell-mimicking nanovesicles potentiate effective cancer immunotherapy, Sci. Adv., 6, 10.1126/sciadv.abd1631
Chen, 2014, Janus particles as artificial antigen-presenting cells for T cell activation, ACS Appl. Mater. Interfaces, 6, 18435, 10.1021/am505510m
Liu, 2020, Heterogeneous polymeric particles encapsulating human T cells for controlled activation, proliferation, and delivery, ACS Appl. Bio Mater., 3, 7357, 10.1021/acsabm.0c00992
Tang, 2018, Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery, Nat. Biotechnol., 36, 707, 10.1038/nbt.4181
Gubin, 2014, Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens, Nature, 515, 577, 10.1038/nature13988
Melero, 2015, Evolving synergistic combinations of targeted immunotherapies to combat cancer, Nat. Rev. Cancer, 15, 457, 10.1038/nrc3973
Kosmides, 2017, Dual targeting nanoparticle stimulates the immune system to inhibit tumor growth, ACS Nano, 11, 5417, 10.1021/acsnano.6b08152
Mi, 2018, A dual immunotherapy nanoparticle improves t-cell activation and cancer immunotherapy, Adv. Mater., 30
Zhang, 2019, An artificial antigen-presenting cell delivering 11 immune molecules expands tumor antigen-specific CTLs in ex vivo and in vivo murine melanoma models, Cancer Immunol. Res., 7, 1188, 10.1158/2326-6066.CIR-18-0881
Huang, 2020, Highly efficient and tumor-selective nanoparticles for dual-targeted immunogene therapy against cancer, Sci. Adv., 6, 10.1126/sciadv.aax5032
Perez Del Rio, 2020, CCL21-loaded 3D hydrogels for T cell expansion and differentiation, Biomaterials, 259, 10.1016/j.biomaterials.2020.120313
Griffith, 1989, In vivo distribution of adoptively transferred indium-111-labeled tumor infiltrating lymphocytes and peripheral blood lymphocytes in patients with metastatic melanoma, J. Natl. Cancer Inst., 81, 1709, 10.1093/jnci/81.22.1709
Schmeichel, 2003, Modeling tissue-specific signaling and organ function in three dimensions, J. Cell Sci., 116, 2377, 10.1242/jcs.00503
Aliperta, 2017, Cryogel-supported stem cell factory for customized sustained release of bispecific antibodies for cancer immunotherapy, Sci. Rep., 7, 42855, 10.1038/srep42855
Hickey, 2019, Engineering an artificial T-cell stimulating matrix for immunotherapy, Adv. Mater., 31
Mattheolabakis, 2015, Hyaluronic acid targeting of CD44 for cancer therapy: from receptor biology to nanomedicine, J. Drug Target., 23, 605, 10.3109/1061186X.2015.1052072
Monette, 2016, Chitosan thermogels for local expansion and delivery of tumor-specific T lymphocytes towards enhanced cancer immunotherapies, Biomaterials, 75, 237, 10.1016/j.biomaterials.2015.10.021
Weiden, 2018, Injectable biomimetic hydrogels as tools for efficient T cell expansion and delivery, Front. Immunol., 9, 2798, 10.3389/fimmu.2018.02798
Guasch, 2017, Integrin-assisted T-cell activation on nanostructured hydrogels, Nano Lett., 17, 6110, 10.1021/acs.nanolett.7b02636
Dang, 2018, Enhanced activation and expansion of T cells using mechanically soft elastomer fibers, Adv. Biosyst., 2, 10.1002/adbi.201700167
Song, 2019, PEGylated and CD47-conjugated nanoellipsoidal artificial antigen-presenting cells minimize phagocytosis and augment anti-tumor T-cell responses, Int. J. Nanomed., 14, 2465, 10.2147/IJN.S195828
Fadel, 2008, Enhanced cellular activation with single walled carbon nanotube bundles presenting antibody stimuli, Nano Lett., 8, 2070, 10.1021/nl080332i
Sunshine, 2014, Particle shape dependence of CD8+ T cell activation by artificial antigen presenting cells, Biomaterials, 35, 269, 10.1016/j.biomaterials.2013.09.050
Meyer, 2015, Biodegradable nanoellipsoidal artificial antigen presenting cells for antigen specific T-cell activation, Small, 11, 1519, 10.1002/smll.201402369
Meyer, 2018, Anisotropic biodegradable lipid coated particles for spatially dynamic protein presentation, Acta Biomater., 72, 228, 10.1016/j.actbio.2018.03.056
Hassan, 2016, Dual stimulation of antigen presenting cells using carbon nanotube-based vaccine delivery system for cancer immunotherapy, Biomaterials, 104, 310, 10.1016/j.biomaterials.2016.07.005
Fadel, 2013, Adsorption of multimeric T cell antigens on carbon nanotubes: effect on protein structure and antigen-specific T cell stimulation, Small, 9, 666, 10.1002/smll.201201684
Fadel, 2014, A carbon nanotube-polymer composite for T-cell therapy, Nat. Nanotechnol., 9, 639, 10.1038/nnano.2014.154
Ye, 2018, Engineering chimeric antigen receptor-T cells for cancer treatment, Mol. Cancer, 17, 32, 10.1186/s12943-018-0814-0
Oelkrug, 2014, Enhancement of T cell recruitment and infiltration into tumours, Clin. Exp. Immunol., 178, 1, 10.1111/cei.12382
Zhan, 2017, Drug-eluting scaffold inhibited in vivo pancreatic tumorigenesis by engaging murine CCR4+CD8+ T cells, Colloids Surf., B, 158, 469, 10.1016/j.colsurfb.2017.07.021
Kwee, 2019, Treating ischemia via recruitment of antigen-specific T cells, Sci. Adv., 5, 10.1126/sciadv.aav6313
Pugliese, 2017, Autoreactive T cells in type 1 diabetes, J. Clin. Invest., 127, 2881, 10.1172/JCI94549
Thelin, 2017, In vivo enrichment of diabetogenic T cells, Diabetes, 66, 2220, 10.2337/db16-0946
Dominguez-Villar, 2018, Regulatory T cells in autoimmune disease, Nat. Immunol., 19, 665, 10.1038/s41590-018-0120-4
Saleh, 2019, Treg-mediated acquired resistance to immune checkpoint inhibitors, Cancer Lett., 457, 168, 10.1016/j.canlet.2019.05.003
González-Navajas, 2021, The Impact of Tregs on the anticancer immunity and the efficacy of immune checkpoint inhibitor therapies, Front. Immunol., 12, 10.3389/fimmu.2021.625783
Josefowicz, 2012, Regulatory T cells: mechanisms of differentiation and function, Annu. Rev. Immunol., 30, 531, 10.1146/annurev.immunol.25.022106.141623
Tivol, 1995, Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4, Immunity, 3, 541, 10.1016/1074-7613(95)90125-6
Liu, 2018, Nanofibrous spongy microspheres to distinctly release mirna and growth factors to enrich regulatory T cells and rescue periodontal bone loss, ACS Nano, 12, 9785, 10.1021/acsnano.7b08976
Fisher, 2019, Treg-inducing microparticles promote donor-specific tolerance in experimental vascularized composite allotransplantation, Proc. Natl. Acad. Sci. U. S. A., 116, 25784, 10.1073/pnas.1910701116
Balmert, 2017, In vivo induction of regulatory T cells promotes allergen tolerance and suppresses allergic contact dermatitis, J. Control. Release, 261, 223, 10.1016/j.jconrel.2017.07.006
Jhunjhunwala, 2012, Controlled release formulations of IL-2, TGF-β1 and rapamycin for the induction of regulatory T cells, J. Control. Release, 159, 78, 10.1016/j.jconrel.2012.01.013
Fisher, 2020, In situ recruitment of regulatory T cells promotes donor-specific tolerance in vascularized composite allotransplantation, Sci. Adv., 6, 10.1126/sciadv.aax8429
Sands, 2020, Tuning cytokines enriches dendritic cells and regulatory T cells in the periodontium, J. Periodontol., 91, 1475, 10.1002/JPER.19-0411
Yao, 2012, Levels of peripheral CD4(+)FoxP3(+) regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancer, Blood, 119, 5688, 10.1182/blood-2011-10-386482
Estrellas, 2018, Biological scaffold–mediated delivery of myostatin inhibitor promotes a regenerative immune response in an animal model of Duchenne muscular dystrophy, J. Biol. Chem., 293, 15594, 10.1074/jbc.RA118.004417
Verbeke, 2017, Multicomponent injectable hydrogels for antigen-specific tolerogenic immune modulation, Adv. Healthcare Mater., 6, 10.1002/adhm.201600773
Majedi, 2018, Cytokine secreting microparticles engineer the fate and the effector functions of T-cells, Adv. Mater., 30, 10.1002/adma.201703178
Kim, 2015, Injectable, spontaneously assembling, inorganic scaffolds modulate immune cells in vivo and increase vaccine efficacy, Nat. Biotechnol., 33, 64, 10.1038/nbt.3071
Dellacherie, 2018, Covalent conjugation of peptide antigen to mesoporous silica rods to enhance cellular responses, Bioconjugate Chem., 29, 733, 10.1021/acs.bioconjchem.7b00656
Sinha, 2019, A 3D macroporous alginate graphene scaffold with an extremely slow release of a loaded cargo for in situ long-term activation of dendritic cells, Adv. Healthcare Mater., 8
Lynn, 2020, Peptide-TLR-7/8a conjugate vaccines chemically programmed for nanoparticle self-assembly enhance CD8 T-cell immunity to tumor antigens, Nat. Biotechnol., 38, 320, 10.1038/s41587-019-0390-x
Blazar, 2012, Advances in graft-versus-host disease biology and therapy, Nat. Rev. Immunol., 12, 443, 10.1038/nri3212
Shah, 2019, An injectable bone marrow-like scaffold enhances T cell immunity after hematopoietic stem cell transplantation, Nat. Biotechnol., 37, 293, 10.1038/s41587-019-0017-2
Srivastava, 2018, Chimeric antigen receptor T cell therapy: challenges to bench-to-bedside efficacy, J Immunol., 200, 459, 10.4049/jimmunol.1701155
Zhao, 2019, Engineered T cell therapy for cancer in the clinic, Front. Immunol., 10, 2250, 10.3389/fimmu.2019.02250
Tsao, 2014, Thermoreversible poly(ethylene glycol)-g-chitosan hydrogel as a therapeutic t lymphocyte depot for localized glioblastoma immunotherapy, Biomacromolecules, 15, 2656, 10.1021/bm500502n
Stephan, 2015, Biopolymer implants enhance the efficacy of adoptive T-cell therapy, Nat. Biotechnol., 33, 97, 10.1038/nbt.3104
Smith, 2017, Biopolymers codelivering engineered T cells and STING agonists can eliminate heterogeneous tumors, J. Clin. Invest., 127, 2176, 10.1172/JCI87624
Ahmadi, 2015, Chitosan based hydrogels: characteristics and pharmaceutical applications, Res. Pharm. Sci., 10, 1
Luo, 2020, Injectable porous microchips with oxygen reservoirs and an immune-niche enhance the efficacy of CAR T cell therapy in solid tumors, ACS Appl. Mater. Interfaces, 12, 56712, 10.1021/acsami.0c15239
Wang, 2020, GD2-specific CAR T cells encapsulated in an injectable hydrogel control retinoblastoma and preserve vision, Nat. Cancer, 1, 990, 10.1038/s43018-020-00119-y
Coon, 2020, Nitinol thin films functionalized with CAR-T cells for the treatment of solid tumours, Nat. Biomed. Eng., 4, 195, 10.1038/s41551-019-0486-0
Hu, 2021, Inhibition of post-surgery tumour recurrence via a hydrogel releasing CAR-T cells and anti-PDL1-conjugated platelets, Nat. Biomed. Eng., 10.1038/s41551-021-00712-1
Hu, 2018, Conjugation of haematopoietic stem cells and platelets decorated with anti-PD-1 antibodies augments anti-leukaemia efficacy, Nat. Biomed. Eng., 2, 831, 10.1038/s41551-018-0310-2
Benjamin, 2020, Genome-edited, donor-derived allogeneic anti-CD19 chimeric antigen receptor T cells in paediatric and adult B-cell acute lymphoblastic leukaemia: results of two phase 1 studies, Lancet, 396, 1885, 10.1016/S0140-6736(20)32334-5
Depil, 2020, 'Off-the-shelf' allogeneic CAR T cells: development and challenges, Nat. Rev. Drug Discovery, 19, 185, 10.1038/s41573-019-0051-2
Perez, 2020, Off-the-shelf allogeneic t cell therapies for cancer: opportunities and challenges using naturally occurring "universal" donor T cells, Front. Immunol., 11, 10.3389/fimmu.2020.583716
Seet, 2017, Generation of mature T cells from human hematopoietic stem and progenitor cells in artificial thymic organoids, Nat. Methods, 14, 521, 10.1038/nmeth.4237
Montel-Hagen, 2020, In vitro recapitulation of murine thymopoiesis from single hematopoietic stem cells, Cell Rep., 33, 10.1016/j.celrep.2020.108320
Asnaghi, 2021, Thymus extracellular matrix-derived scaffolds support graft-resident thymopoiesis and long-term in vitro culture of adult thymic epithelial cells, Adv. Funct. Mater., 31, 10.1002/adfm.202010747
Campinoti, 2020, Reconstitution of a functional human thymus by postnatal stromal progenitor cells and natural whole-organ scaffolds, Nat. Commun., 11, 6372, 10.1038/s41467-020-20082-7
Shevyrev, 2019, Treg heterogeneity, function, and homeostasis, Front. Immunol., 10, 3100, 10.3389/fimmu.2019.03100
Ikebuchi, 2019, Functional phenotypic diversity of regulatory T cells remaining in inflamed skin, Front. Immunol., 10, 1098, 10.3389/fimmu.2019.01098
MacIver, 2013, Metabolic regulation of T lymphocytes, Annu. Rev. Immunol., 31, 259, 10.1146/annurev-immunol-032712-095956
Shi, 2019, Metabolic control of treg cell stability, plasticity, and tissue-specific heterogeneity, Front. Immunol., 10, 2716, 10.3389/fimmu.2019.02716
Hippen, 2011, Massive ex vivo expansion of human natural regulatory T cells (T(regs)) with minimal loss of in vivo functional activity, Sci. Transl. Med., 3, 10.1126/scitranslmed.3001809
Rurik, 2022, CAR T cells produced in vivo to treat cardiac injury, Science, 375, 91, 10.1126/science.abm0594
Parayath, 2020, In vitro-transcribed antigen receptor mRNA nanocarriers for transient expression in circulating T cells in vivo, Nat. Commun., 11, 6080, 10.1038/s41467-020-19486-2
Zhang, 2019, Decoy oligodeoxynucleotides, polysaccharides, and targeted peptide-functionalized gold nanorods for the combined treatment of rheumatoid arthritis, Adv. Healthcare Mater., 8, 10.1002/adhm.201970074
Aguado, 2018, Engineering precision biomaterials for personalized medicine, Sci. Transl. Med., 10, 10.1126/scitranslmed.aam8645