Abstract
Exciting developments in cancer nanomedicine include the engineering of nanocarriers to deliver drugs locally to tumors, increasing efficacy and reducing off-target toxicity associated with chemotherapies. Despite nanocarrier advances, metastatic cancer remains challenging to treat due to barriers that prevent nanoparticles from gaining access to remote, dispersed, and poorly vascularized metastatic tumors. Instead of relying on nanoparticles to directly destroy every tumor cell, immunotherapeutic approaches target immune cells to train them to recognize and destroy tumor cells, which, due to the amplification and specificity of an adaptive immune response, may be a more effective approach to treating metastatic cancer. One novel technology for cancer immunotherapy is the artificial antigen presenting cell (aAPC), a micro- or nanoparticle-based system that mimics an antigen presenting cell by presenting important signal proteins to T cells to activate them against cancer. Signal 1 molecules target the T cell receptor and facilitate antigen recognition by T cells, signal 2 molecules provide costimulation essential for T cell activation, and signal 3 consists of secreted cues that further stimulate T cells. Classic microscale aAPCs present signal 1 and 2 molecules on their surface, and biodegradable polymeric aAPCs offer the additional capability of releasing signal 3 cytokines and costimulatory molecules that modulate the T cell response. Although particles of approximately 5–10 microns in diameter may be considered the optimal size of an aAPC for ex vivo cellular expansion, nanoscale aAPCs have demonstrated superior in vivo pharmacokinetic properties and are more suitable for systemic injection. As sufficient surface contact between T cells and aAPCs is essential for activation, nano-aAPCs with microscale contact surface areas have been created through engineering approaches such as shape manipulation and nanoparticle clustering. These design strategies have demonstrated greatly enhanced efficacy of nano-aAPCs, endowing nano-aAPCs with the potential to be among the next generation of cancer nanomedicines.
Keywords: nanomedicine, aAPC, immunoengineering, cancer, polymer
Graphical Abstract
Introduction
Recent decades have seen major advances in the use of nanomedicine to treat cancer. Cancer nanomedicine is the application of nanoscale tools, especially nanoscale particulates, to diagnose, treat, and prevent the disease (1). Current cancer nanomedicine therapeutics utilize nanoparticles engineered to deliver drug cargos locally to tumor sites, reducing negative systemic side effects, such as those associated with chemotherapies (2). These nanocarriers can encapsulate and deliver poorly soluble drugs, stabilize drugs, and slow their degradation and elimination (3). These vectors also take advantage of the enhanced permeation and retention (EPR) of nanoparticles in the leaky vasculature associated with tumors, allowing for passive tumor targeting and a decrease in off-target toxicity (2). In addition to passive targeting, nanoparticles can be actively targeted to tumors through the conjugation of ligands including monoclonal antibodies, peptides, antibody fragments, or small molecules that bind to specific markers on the surface of tumor cells (2, 4, 5). Nanocarriers can be fabricated from various biomaterials, including ones that are lipid-based (3, 6), albumin-bound (7), inorganic (2), or polymeric (8–12), the latter of which are advantageous because they can be constructed from biocompatible, biodegradable materials that encapsulate and release drugs in a controlled manner.
Despite nanocarrier advances, metastatic cancer is still considered largely incurable and is responsible for the vast majority of cancer deaths. Metastases are often poorly vascularized compared to primary tumors, and cannot be effectively accessed by nanoparticles via the EPR effect (13). Additionally, metastases can present in multiple organ environments, each with a unique set of challenges that must be considered when designing particles to target and penetrate the tumor (13, 14). To potentially cure metastatic cancer and prevent recurrence, a nanocarrier therapy must overcome these barriers and deliver its drug cargo to kill every tumor cell. Immunotherapies may be a more effective strategy than traditional nanomedicines because they harness the power of the host’s immune system to fight the tumor. Instead of relying on nanoparticles to destroy every tumor cell, immunotherapeutic approaches activate the adaptive immune system against tumor-specific antigens, leading immune cells to mount an amplified response to recognize and destroy tumor cells (15). One promising technology for cancer immunotherapy is the artificial antigen presenting cell (aAPC), a micro- or nanoparticle-based biomimetic system that functions like a natural antigen presenting cell (APC) to activate the adaptive immune system against cancer. Through their ease of production and their ability to efficiently activate and expand antigen-specific T cell populations, aAPCs show exciting potential as an “off-the-shelf” cancer immunotherapy (16).
The aAPC: A Biomimetic Platform for Cancer Immunotherapy
aAPCs are synthetic constructs that mimic the natural process of T cell activation by biological APCs through the presentation of essential T cell-stimulating signal proteins conjugated to their surface. When aAPCs interact with T cells, these signal proteins activate the T cells and modulate their response. Cellular aAPC platforms to expand antigen-specific CD8+ T cells for adoptive T cell therapies are under clinical investigations to treat a variety of cancers including advanced melanoma (17), but scale-up is expensive and labor-intensive. Acellular aAPC platforms are more easily produced, can be stored as an off-the-shelf therapy, and can be readily adapted to accommodate a variety of antigen specificities and surface ligands. As synthetic systems, they also eliminate the risk of tumorgenicity associated with cellular aAPCs and are unaffected by the immunosuppressive tumor microenvironment (18).
Signals 1, 2, and 3: Antigen presentation, Costimulation, and Cytokine Release
Classic aAPCs present signal 1 and 2 molecules on their surface that are critical for T cell activation (Fig. 1). Signal 1 is used in APC/T cell recognition and provides antigen-specificity. The majority of nonspecific aAPCs bear an antibody that stimulates the CD3 T cell co-receptor, which associates with the TCR complex to produce an activation signal within the T cell. Nonspecific targeting with anti-CD3 is often employed in aAPC systems, but antigen specificity is necessary in a therapeutic setting for efficient expansion of effector T cells against tumor-specific antigens (19). Antigen-specific signal 1 can be provided by MHC single chain complexes, but MHC multimers offer more efficient T cell activation and peptide-loading flexibility (19, 20). Signal 1 density on the aAPC surface affects T cell activation. Matic et al. demonstrated that an anti-CD3 density of 1000 proteins/um2 increased T cell proliferation and IL-2 production compared to 120 and 60 proteins/um2 (21).
Figure 1.
Strategies for aAPC design. A) A classic micro-aAPC with surface-bound signal 1 (anti-CD3, MHC multimer, etc.) and signal 2 (anti-CD28, anti-4-1BB, etc.) molecules. B) Newer aAPC systems are constructed from biodegradable materials capable of releasing costimulatory and signal 3 cytokines (IL-12, IFN-α/β, etc.). C) Nano-aAPCs are less efficient T cell activators, but may outperform micro-aAPCs in vivo due to their transport properties. D) Recent findings suggest that ellipsoidal nano-aAPCs activate T cells more efficiently compared to spherical nano-aAPCs due to increased contact surface area. E) Magnetic field-induced clustering of paramagnetic nano-aAPCs has been shown to increase T cell induction and expansion.
Signal 2 is a costimulatory signal required for T cell activation. Most aAPC platforms (Fig. 1A) utilize an antibody against CD28 as a costimulatory molecule, although 4-1BB can also stimulate antigen-specific T cells in the absence of CD28 costimulation (22, 23). Interestingly, a 75:25 blend of anti-4-1BB and anti-CD28 on the surface of polystyrene micro-aAPCs also presenting an HLA MHC Class I molecule as Signal 1 led to a five-fold increase of antigen-specific CD8+ T cells compared to each individual signal (24).
In addition to the type of costimulatory ligand, the strength and density of costimulation affect the nature of cell activation (25). Stimulation of CD4+ cells with anti-CD28, which has a greater affinity for the CD28 receptor than natural costimulatory ligands B7-1 and B7-2, was shown to contribute to a Th1 phenotype (26). Meanwhile, B7-1 costimulation led to a Th2 response in CD4+ T cells (26) and was insufficient to sustain a CD8+ T cell response (27). CD8+ cells were shown to require a higher density of costimulatory molecules than CD4+ cells to achieve the same level of activation (27). In a liposomal aAPC, a density of 2000 molecules/um2 was found to be optimal for stimulation with a 100:1 liposome to target cell ratio (28).
Although not necessary for activation, secreted cytokines provide a third signal that influences T cell effector phenotype (29). Biodegradable polymeric aAPCs are capable of releasing signal 3 cytokines and costimulatory molecules (Fig. 1B). Steenblock et al. showed that release of IL-2 from PLGA aAPCs with stable surface ligand presentation resulted in a 3–4-fold increase in T cell expansion compared to an equivalent amount of exogenous IL-2, demonstrating the enhancement in effective concentration caused by local delivery and close proximity between a drug releasing particle and a cell (30). These aAPC systems have been reproduced and shown to be effective in multiple studies (31, 32). Zhang et al. demonstrated co-release of IL-2 and anti-CTLA-4 from PLGA micro-aAPCs presenting surface-bound MHC dimer and anti-CD28 (33). These aAPCs significantly slowed tumor growth and enhanced survival in a murine melanoma model over a combination of aAPCs releasing each signal individually, and non-releasing aAPCs with IL-2 and anti-CLTA-4 injected systemically (33).
Micro-aAPCs for Cancer Immunotherapy
aAPC particles with a diameter of 5–10 microns, approximately the same size as biological APCs, have been shown to be most efficient at T cell stimulation for tumor immunotherapy (34). In one study, Mescher found that particles at least 4–5 microns in diameter, a biomimetic length scale, were necessary to activate CD8+ T cells. Equivalent activation could not be reached with increased doses of smaller particles (35). Similarly, Steenblock et al. demonstrated that micro-aAPCs induced a 3-fold increase in CD8+ T cell IL-2 production compared to nano-aAPCs bearing the same protein dose (34). Liposomal (36, 37), paramagnetic (38), and polymeric materials (24, 34, 39, 40) presenting a variety of signal 1 and 2 molecules have all been used to successfully expand functional CD8+ T cells in vitro. In one example, Lu et al. utilized polystyrene beads coated with an MHC class I dimer, anti-CD28, 4-1BBL, and CD83 molecules to expand CD8+ T cells ex vivo for adoptive transfer into murine melanoma and pulmonary metastasis models. Adoptively transferred antigen-specific CD8+ cells led to complete regression of established B16 melanoma tumors in a model and significant tumor reduction in the murine metastasis model (41). In an early example of aAPC in vivo efficacy, Shalaby et al. showed that a combination of poly-glycolic acid (PGA) microparticles with surface bound anti-CD3 and anti-CD28 and PGA particles releasing granulocyte macrophage colony-stimulating factor (GM-CSF) from their surface prevented tumor implantation in 100% of mice, as well as complete tumor regression in 57% of mice with established fibrosarcoma tumors (40). Shen et al. demonstrated that polystyrene aAPCs bearing MHC class 1 tetramer and a combination of anti-CD28 and anti-4-1BB on their surface inhibited tumor growth and delayed tumor progression in mice with established B16 melanoma tumors (42). More recently, biodegradable PLGA aAPCs administered in vivo have been shown to prevent implantation, reduce tumor burden, and prolong survival in murine melanoma models (33, 43, 44).
Nano-aAPCs for Cancer Immunotherapy
Microparticulate, and to a lesser extent, nanoparticulate systems (45) have successfully expanded and activated T cells in vitro. However, nanoscale aAPCs (Fig. 1C) may be better suited for in vivo administration than micro-scale aAPCs, due to improved transport properties and a reduced risk of embolism (18). Several nano-aAPC systems have outperformed micro-aAPCs at stimulating antigen-specific CD8+ cells in vivo, their success likely linked to superior biodistribution and drainage properties (45, 46). Despite these successes, nanoscale aAPCs have a much smaller surface area for contact with T cells and are recognized as generally less efficient T cell activators compared to microscale aAPCs. Due to the fact that sufficient contact between the T cell and aAPC is essential for activation, nano-aAPCs with microscale contact surface areas have been created through strategies including shape manipulation (Fig. 1D) and nanoparticle clustering (Fig. 1E).
Anisotropic Particle Shape
Nonspherical, anisotropic nanoparticles can possess a micron length scale radius of curvature on a long axis to approximate the interfacial geometry of micro-aAPCs (Fig. 2A), and display enhanced pharmacokinetic properties compared to spherical particles (47, 48). Ellipsoidal particles demonstrated reduced non-specific uptake (49), increased specific uptake, and enhanced cancer cell killing (50). Compared to spherical and flattened disc-shaped particles, ellipsoidal particles showed the most efficient particle attachment and lowest in vitro internalization rates (51). Sunshine et al. showed that ellipsoidal micro-aAPCs had more frequent interactions with T cells and greater contact area of interaction than corresponding spherical aAPCs and the ellipsoidal aAPCs also led to improved anti-cancer T cell activity in vivo in a B16 melanoma mouse model compared to spherical aAPCs (43). Using the same materials and film stretching method, Meyer et al. created ellipsoidal PLGA nano-aAPCs presenting an MHC Class 1 Ig dimer signal 1 protein and anti-CD28 as signal 2 (48). In vitro, 14% of ellipsoidal nano-aAPCs were phagocytosed by macrophages compared to 78% of spherical nano-aAPCs. When administered intravenously, higher concentrations of ellipsoidal particles remained in the bloodstream for an hour post-injection and were better dispersed (Fig. 2B). Following intravenous administration with adoptively transferred PMEL CD8+ cells, ellipsoidal nano-aAPCs led to 2-fold greater CD8+ T cell expansion in a B16 melanoma model (Fig. 2C). Overall, ellipsoidal nano-aAPCs demonstrated superior immunogenicity and pharmacokinetic properties in vivo compared to spherical nanoparticles (48).
Figure 2.
Ellipsoidal and magnetically clustered nano-aAPC enhance antigen-specific T cell activation and expansion in vivo. A) Ellipsoidal nanoparticles were created by mechanical stretching. (Scale bar is 500 nm). B) Fluorescent ellipsoidal nano-aAPCs circulate with a longer half-life in the bloodstream compared to spherical nano-APCs. C) Ellipsoidal nano-aAPCs led to a statistically significant increase in the percentage of antigen-specific T cells in vivo compared to spherical nano-aAPCs. Reproduced with permission from Small (48). D) Schematic of magnetic field-induced clustering of TCRs bound to paramagnetic nanoparticles. E) T cells stimulated with nano-aAPCs in a magnetic field and adoptively transferred were present at higher frequencies seven days post-injection compared to controls. F) Treatment with magnetically enhanced T cells led to increased survival when T cells and particles were adoptively transferred on Day 10. Reproduced with permission from ACS Nano <http://pubs.acs.org/doi/10.1021/nn405520d> (52), further permissions related to the material excerpted should be directed to the ACS.
Magnetic Field-Induced Nanoparticle Clustering
Another method to increase the surface contact area between nano-aAPCs and T cells and strengthen activation is through magnetic-field induced nanoparticle clustering (Fig. 2D) (52). Application of an external magnetic field has been shown to induce magnetization and aggregation of superparamagnetic particles and resultant clustering of nanoparticle-bound surface receptors on target cells (53, 54). When T cells interact with APCs, MHC-TCR complexes aggregate at the contact site to form an immunological synapse, which strengthens signaling and subsequent T cell activation. aAPCs with pre-clustered MHC complexes on their surface improved T cell activation compared to those with a uniformly distributed MHC complexes (37). Perica et al. utilized iron-dextran nanoparticles directly conjugated with surface-bound MHC Ig dimer signal 1 and anti-CD28 signal 2 to generate reproducible magnetic aAPCs that have also been used in other studies (45). They demonstrated that nano-clustering in response to an applied magnetic field led to a two-fold increase in TCR/CD3 aggregate size and a two-fold decrease in the number of TCR clusters per T cell compared to a control group with no applied magnetic field (52). The magnetically clustered nano-aAPCs induced 15-fold antigen-specific expansion of CD8+ T cells compared to non-clustered aAPCs (Fig. 2E). Adoptive transfer of magnetically enhanced T cells in mice expressing B16 melanoma tumors significantly reduced tumor burden, prolonged survival, and led to complete tumor regression in 50% of treated mice compared with no treatment or non-enhanced T cell controls (52) (Fig. 2F).
Conclusions and Future Perspective
Despite ongoing nanocarrier development, there are still many challenges associated with the local delivery of nanotherapeutics to treat cancer, particularly in advanced stages. aAPCs are a promising technology that can allow a host to generate an immune response to fight cancer systemically. Micro, and to a lesser extent, nano-aAPCs have shown success in expanding antigen-specific T cell populations ex vivo and successful adoptive transfer into murine cancer models. Nano-aAPCs have the potential for antigen-specific T cell stimulation in vivo and could become a translatable, off-the-shelf cancer therapy without the need for adoptive T cell transfer. In the design of future nano-aAPCs, special consideration must be given to the type, density, organization, and mobility of signals presented on the surface, as well as the encapsulation and controlled release of cytokines from the particles in vivo. Engineering a biomimetic length scale for the aAPC-T cell contact interface is particularly important when designing aAPCs on the nanoscale. The next generation of nano-aAPCs may utilize strategies such as manipulation of physical shape and aspect ratio and application of external triggers to facilitate more accurate approximation of interfacial geometry between biological APCs and T cells. Besides length-scale considerations, other physical and biological properties of the immunological synapse must be optimized in nano-aAPC systems. In addition to particle shape and aspect ratio, parameters such as particle rigidity may be tuned to in future designs to exercise more control over aAPC-T cell interactions. The biological nature of the immunological synapse can be recapitulated through surface engineering strategies such as patterning signal proteins in precise spatial arrangements, or coating aAPCs with supported lipid bilayers to allow signal protein and receptor reorganization in response to T cell contact. Future nano-aAPC systems may benefit from a combination of optimized physical and biological parameters that enhance their biomimicry and efficacy. The use of nano-aAPCs as a biomimetic platform for cancer immunotherapy has strong potential to contribute to the advancement of cancer nanomedicine.
HIGHLIGHTS.
Artificial antigen presenting cells (aAPC) are particle-based systems that mimic an antigen presenting cell by presenting signal 1 and 2 molecules on their surface to activate T cells.
Nanoscale aAPCs can have desirable pharmacokinetic properties that make them suitable for systemic injection.
A micron sized length scale of contact between T cells and aAPCs is desirable for T cell activation.
Nano-aAPCs with microscale contact surface areas have been created through engineering approaches such as shape manipulation and nanoparticle clustering.
The use of nano-aAPCs as a biomimetic platform for cancer immunotherapy has strong potential to contribute to the advancement of cancer nanomedicine.
Acknowledgments
The authors thank the NIH for support (NIH R01EB022148) as well as the Bloomberg-Kimmel Institute for Cancer Immunotherapy. KRR thanks the NSF for a Graduate Research Fellowship.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Freitas RA., Jr What is nanomedicine? Nanomedicine. 2005;1(1):2–9. doi: 10.1016/j.nano.2004.11.003. PubMed PMID: 17292052. [DOI] [PubMed] [Google Scholar]
- 2.Wang AZ, Langer R, Farokhzad OC. Nanoparticle delivery of cancer drugs. Annu Rev Med. 2012;63:185–98. doi: 10.1146/annurev-med-040210-162544. PubMed PMID: 21888516. [DOI] [PubMed] [Google Scholar]
- 3.Wicki A, Witzigmann D, Balasubramanian V, Huwyler J. Nanomedicine in cancer therapy: challenges, opportunities, and clinical applications. J Control Release. 2015;200:138–57. doi: 10.1016/j.jconrel.2014.12.030. PubMed PMID: 25545217. [DOI] [PubMed] [Google Scholar]
- 4.Wang AZ, Gu F, Zhang L, Chan JM, Radovic-Moreno A, Shaikh MR, Farokhzad OC. Biofunctionalized targeted nanoparticles for therapeutic applications. Expert Opin Biol Ther. 2008;8(8):1063–70. doi: 10.1517/14712598.8.8.1063. PubMed PMID: 18613759; PMCID: PMC3515649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Park JH, Lee S, Kim J-H, Park K, Kim K, Kwon IC. Polymeric nanomedicine for cancer therapy. Progress in Polymer Science. 2008;33(1):113–37. doi: 10.1016/j.progpolymsci.2007.09.003. [DOI] [Google Scholar]
- 6.Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov. 2005;4(2):145–60. doi: 10.1038/nrd1632. PubMed PMID: 15688077. [DOI] [PubMed] [Google Scholar]
- 7.Hawkins MJ, Soon-Shiong P, Desai N. Protein nanoparticles as drug carriers in clinical medicine. Adv Drug Deliv Rev. 2008;60(8):876–85. doi: 10.1016/j.addr.2007.08.044. PubMed PMID: 18423779. [DOI] [PubMed] [Google Scholar]
- 8.van der Meel R, Lammers T, Hennink WE. Cancer nanomedicines: oversold or underappreciated? Expert Opin Drug Deliv. 2017;14(1):1–5. doi: 10.1080/17425247.2017.1262346. PubMed PMID: 27852113; PMCID: PMC5404718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Baker JR., Jr Dendrimer-based nanoparticles for cancer therapy. Hematology Am Soc Hematol Educ Program. 2009:708–19. doi: 10.1182/asheducation-2009.1.708. PubMed PMID: 20008257. [DOI] [PubMed] [Google Scholar]
- 10.van Elk M, Murphy BP, Eufrasio-da-Silva T, O'Reilly DP, Vermonden T, Hennink WE, Duffy GP, Ruiz-Hernandez E. Nanomedicines for advanced cancer treatments: Transitioning towards responsive systems. Int J Pharm. 2016;515(1–2):132–64. doi: 10.1016/j.ijpharm.2016.10.013. Epub 2016/11/05. PubMed PMID: 27725268. [DOI] [PubMed] [Google Scholar]
- 11.Banik BL, Fattahi P, Brown JL. Polymeric nanoparticles: the future of nanomedicine. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2016;8(2):271–99. doi: 10.1002/wnan.1364. Epub 2015/09/01. PubMed PMID: 26314803. [DOI] [PubMed] [Google Scholar]
- 12.Plummer R, Wilson RH, Calvert H, Boddy AV, Griffin M, Sludden J, Tilby MJ, Eatock M, Pearson DG, Ottley CJ, Matsumura Y, Kataoka K, Nishiya T. A Phase I clinical study of cisplatin-incorporated polymeric micelles (NC-6004) in patients with solid tumours. Br J Cancer. 2011;104(4):593–8. doi: 10.1038/bjc.2011.6. Epub 2011/02/03. PubMed PMID: 21285987; PMCID: PMC3049602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Schroeder A, Heller DA, Winslow MM, Dahlman JE, Pratt GW, Langer R, Jacks T, Anderson DG. Treating metastatic cancer with nanotechnology. Nat Rev Cancer. 2011;12(1):39–50. doi: 10.1038/nrc3180. PubMed PMID: 22193407. [DOI] [PubMed] [Google Scholar]
- 14.Truffi M, Sorrentino L, Mazzucchelli S, Fiandra L, Corsi F. What Is the Role of Nanotechnology in Diagnosis and Treatment of Metastatic Breast Cancer? Promising Scenarios for the Near Future. Journal of Nanomaterials. 2016;2016:16. doi: 10.1155/2016/5436458. [DOI] [Google Scholar]
- 15.Tong R, Kohane DS. New Strategies in Cancer Nanomedicine. Annu Rev Pharmacol Toxicol. 2016;56:41–57. doi: 10.1146/annurev-pharmtox-010715-103456. Epub 2015/10/31. PubMed PMID: 26514197. [DOI] [PubMed] [Google Scholar]
- 16.Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature. 2011;480(7378):480–9. doi: 10.1038/nature10673. Epub 2011/12/24. PubMed PMID: 22193102; PMCID: PMC3967235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Butler MO, Hirano N. Human cell-based artificial antigen-presenting cells for cancer immunotherapy. Immunol Rev. 2014;257(1):191–209. doi: 10.1111/imr.12129. PubMed PMID: 24329798; PMCID: PMC3869003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Steenblock ER, Wrzesinski SH, Flavell RA, Fahmy TM. Antigen presentation on artificial acellular substrates: modular systems for flexible, adaptable immunotherapy. Expert Opin Biol Ther. 2009;9(4):451–64. doi: 10.1517/14712590902849216. PubMed PMID: 19344282. [DOI] [PubMed] [Google Scholar]
- 19.Oelke M, Krueger C, Giuntoli RL, 2nd, Schneck JP. Artificial antigen-presenting cells: artificial solutions for real diseases. Trends Mol Med. 2005;11(9):412–20. doi: 10.1016/j.molmed.2005.07.005. Epub 2005/08/17. PubMed PMID: 16103011. [DOI] [PubMed] [Google Scholar]
- 20.Maus MV, Riley JL, Kwok WW, Nepom GT, June CH. HLA tetramer-based artificial antigen-presenting cells for stimulation of CD4+ T cells. Clin Immunol. 2003;106(1):16–22. doi: 10.1016/s1521-6616(02)00017-7. Epub 2003/02/14. PubMed PMID: 12584046. [DOI] [PubMed] [Google Scholar]
- 21.Matic J, Deeg J, Scheffold A, Goldstein I, Spatz JP. Fine tuning and efficient T cell activation with stimulatory aCD3 nanoarrays. Nano Lett. 2013;13(11):5090–7. doi: 10.1021/nl4022623. Epub 2013/10/12. PubMed PMID: 24111628; PMCID: PMC3834297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chacon JA, Wu RC, Sukhumalchandra P, Molldrem JJ, Sarnaik A, Pilon-Thomas S, Weber J, Hwu P, Radvanyi L. Co-stimulation through 4-1BB/CD137 improves the expansion and function of CD8(+) melanoma tumor-infiltrating lymphocytes for adoptive T-cell therapy. PLoS One. 2013;8(4):e60031. doi: 10.1371/journal.pone.0060031. Epub 2013/04/06. PubMed PMID: 23560068; PMCID: PMC3613355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Zhang H, Snyder KM, Suhoski MM, Maus MV, Kapoor V, June CH, Mackall CL. 4-1BB is superior to CD28 costimulation for generating CD8+ cytotoxic lymphocytes for adoptive immunotherapy. J Immunol. 2007;179(7):4910–8. doi: 10.4049/jimmunol.179.7.4910. Epub 2007/09/20. PubMed PMID: 17878391; PMCID: PMC3809056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Rudolf D, Silberzahn T, Walter S, Maurer D, Engelhard J, Wernet D, Buhring HJ, Jung G, Kwon BS, Rammensee HG, Stevanovic S. Potent costimulation of human CD8 T cells by anti-4-1BB and anti-CD28 on synthetic artificial antigen presenting cells. Cancer Immunol Immunother. 2008;57(2):175–83. doi: 10.1007/s00262-007-0360-x. PubMed PMID: 17657490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Balmert SC, Little SR. Biomimetic delivery with micro- and nanoparticles. Adv Mater. 2012;24(28):3757–78. doi: 10.1002/adma.201200224. Epub 2012/04/25. PubMed PMID: 22528985; PMCID: PMC3627374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Broeren CP, Gray GS, Carreno BM, June CH. Costimulation light: activation of CD4+ T cells with CD80 or CD86 rather than anti-CD28 leads to a Th2 cytokine profile. J Immunol. 2000;165(12):6908–14. doi: 10.4049/jimmunol.165.12.6908. Epub 2000/12/20. PubMed PMID:11120816. [DOI] [PubMed] [Google Scholar]
- 27.Deeths MJ, Mescher MF. B7-1-dependent co-stimulation results in qualitatively and quantitatively different responses by CD4+ and CD8+ T cells. Eur J Immunol. 1997;27(3):598–608. doi: 10.1002/eji.1830270305. Epub 1997/03/01. PubMed PMID: 9079798. [DOI] [PubMed] [Google Scholar]
- 28.Engelhard VH, Strominger JL, Mescher M, Burakoff S. Induction of secondary cytotoxic T lymphocytes by purified HLA-A and HLA-B antigens reconstituted into phospholipid vesicles. Proc Natl Acad Sci U S A. 1978;75(11):5688–91. doi: 10.1073/pnas.75.11.5688. Epub 1978/11/01. PubMed PMID: 310125; PMCID: PMC393033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Curtsinger JM, Schmidt CS, Mondino A, Lins DC, Kedl RM, Jenkins MK, Mescher MF. Inflammatory cytokines provide a third signal for activation of naive CD4+ and CD8+ T cells. J Immunol. 1999;162(6):3256–62. Epub 1999/03/27. PubMed PMID: 10092777. [PubMed] [Google Scholar]
- 30.Steenblock ER, Fadel T, Labowsky M, Pober JS, Fahmy TM. An artificial antigen-presenting cell with paracrine delivery of IL-2 impacts the magnitude and direction of the T cell response. J Biol Chem. 2011;286(40):34883–92. doi: 10.1074/jbc.M111.276329. Epub 2011/08/19. PubMed PMID: 21849500; PMCID: PMC3186438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Fahmy TM, Samstein RM, Harness CC, Mark Saltzman W. Surface modification of biodegradable polyesters with fatty acid conjugates for improved drug targeting. Biomaterials. 2005;26(28):5727–36. doi: 10.1016/j.biomaterials.2005.02.025. Epub 2005/05/10. PubMed PMID: 15878378. [DOI] [PubMed] [Google Scholar]
- 32.Park J, Mattessich T, Jay SM, Agawu A, Saltzman WM, Fahmy TM. Enhancement of surface ligand display on PLGA nanoparticles with amphiphilic ligand conjugates. J Control Release. 2011;156(1):109–15. doi: 10.1016/j.jconrel.2011.06.025. Epub 2011/07/05. PubMed PMID: 21723893; PMCID: PMC3800156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhang L, Wang L, Shahzad KA, Xu T, Wan X, Pei W, Shen C. Paracrine release of IL-2 and anti-CTLA-4 enhances the ability of artificial polymer antigen-presenting cells to expand antigen-specific T cells and inhibit tumor growth in a mouse model. Cancer Immunol Immunother. 2017 doi: 10.1007/s00262-017-2016-9. Epub 2017/05/16. PubMed PMID: 28501941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Steenblock ER, Fahmy TM. A comprehensive platform for ex vivo T-cell expansion based on biodegradable polymeric artificial antigen-presenting cells. Mol Ther. 2008;16(4):765–72. doi: 10.1038/mt.2008.11. Epub 2008/03/13. PubMed PMID:18334990. [DOI] [PubMed] [Google Scholar]
- 35.Mescher MF. Surface contact requirements for activation of cytotoxic T lymphocytes. J Immunol. 1992;149(7):2402–5. Epub 1992/10/01. PubMed PMID:1527386. [PubMed] [Google Scholar]
- 36.Prakken B, Wauben M, Genini D, Samodal R, Barnett J, Mendivil A, Leoni L, Albani S. Artificial antigen-presenting cells as a tool to exploit the immune 'synapse'. Nat Med. 2000;6(12):1406–10. doi: 10.1038/82231. PubMed PMID: 11100129. [DOI] [PubMed] [Google Scholar]
- 37.Giannoni F, Barnett J, Bi K, Samodal R, Lanza P, Marchese P, Billetta R, Vita R, Klein MR, Prakken B, Kwok WW, Sercarz E, Altman A, Albani S. Clustering of T cell ligands on artificial APC membranes influences T cell activation and protein kinase C theta translocation to the T cell plasma membrane. J Immunol. 2005;174(6):3204–11. doi: 10.4049/jimmunol.174.6.3204. PubMed PMID: 15749850. [DOI] [PubMed] [Google Scholar]
- 38.Oelke M, Maus MV, Didiano D, June CH, Mackensen A, Schneck JP. Ex vivo induction and expansion of antigen-specific cytotoxic T cells by HLA-Ig-coated artificial antigen-presenting cells. Nat. Med. 2003;9(5):619–24. doi: 10.1038/nm869. PubMed PMID: 12704385. [DOI] [PubMed] [Google Scholar]
- 39.Tham EL, Jensen PL, Mescher MF. Activation of antigen-specific T cells by artificial cell constructs having immobilized multimeric peptide-class I complexes and recombinant B7-Fc proteins. J Immunol Methods. 2001;249(1–2):111–9. doi: 10.1016/s0022-1759(00)00335-5. PubMed PMID:11226469. [DOI] [PubMed] [Google Scholar]
- 40.Shalaby WS, Yeh H, Woo E, Corbett JT, Gray H, June CH, Shalaby SW. Absorbable microparticulate cation exchanger for immunotherapeutic delivery. J Biomed Mater Res B Appl Biomater. 2004;69(2):173–82. doi: 10.1002/jbm.b.20040. PubMed PMID:15116407. [DOI] [PubMed] [Google Scholar]
- 41.Lu X, Jiang X, Liu R, Zhao H, Liang Z. Adoptive transfer of pTRP2-specific CTLs expanding by bead-based artificial antigen-presenting cells mediates anti-melanoma response. Cancer Lett. 2008;271(1):129–39. doi: 10.1016/j.canlet.2008.05.049. PubMed PMID: 18621475. [DOI] [PubMed] [Google Scholar]
- 42.Shen C, Cheng K, Miao S, Wang W, He Y, Meng F, Zhang J. Latex bead-based artificial antigen-presenting cells induce tumor-specific CTL responses in the native T-cell repertoires and inhibit tumor growth. Immunol Lett. 2013;150(1–2):1–11. doi: 10.1016/j.imlet.2013.01.003. Epub 2013/01/19. PubMed PMID: 23328744. [DOI] [PubMed] [Google Scholar]
- 43.Sunshine JC, Perica K, Schneck JP, Green JJ. Particle shape dependence of CD8+ T cell activation by artificial antigen presenting cells. Biomaterials. 2014;35(1):269–77. doi: 10.1016/j.biomaterials.2013.09.050. Epub 2013/10/09. PubMed PMID: 24099710; PMCID: PMC3902087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kosmides AK, Meyer RA, Hickey JW, Aje K, Cheung KN, Green JJ, Schneck JP. Biomimetic biodegradable artificial antigen presenting cells synergize with PD-1 blockade to treat melanoma. Biomaterials. 2017;118:16–26. doi: 10.1016/j.biomaterials.2016.11.038. Epub 2016/12/13. PubMed PMID: 27940380; PMCID: PMC5207804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Perica K, De Leon Medero A, Durai M, Chiu YL, Bieler JG, Sibener L, Niemoller M, Assenmacher M, Richter A, Edidin M, Oelke M, Schneck J. Nanoscale artificial antigen presenting cells for T cell immunotherapy. Nanomedicine. 2014;10(1):119–29. doi: 10.1016/j.nano.2013.06.015. Epub 2013/07/31. PubMed PMID: 23891987; PMCID: PMC4114774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Fifis T, Gamvrellis A, Crimeen-Irwin B, Pietersz GA, Li J, Mottram PL, McKenzie IF, Plebanski M. Size-dependent immunogenicity: therapeutic and protective properties of nano-vaccines against tumors. J Immunol. 2004;173(5):3148–54. doi: 10.4049/jimmunol.173.5.3148. Epub 2004/08/24. PubMed PMID: 15322175. [DOI] [PubMed] [Google Scholar]
- 47.Sunshine JC, Green JJ. Nanoengineering approaches to the design of artificial antigen-presenting cells. Nanomedicine (Lond) 2013;8(7):1173–89. doi: 10.2217/nnm.13.98. Epub 2013/07/11. PubMed PMID: 23837856; PMCID: PMC3951141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Meyer RA, Sunshine JC, Perica K, Kosmides AK, Aje K, Schneck JP, Green JJ. Biodegradable nanoellipsoidal artificial antigen presenting cells for antigen specific T-cell activation. Small. 2015;11(13):1519–25. doi: 10.1002/smll.201402369. Epub 2015/02/03. PubMed PMID: 25641795; PMCID: PMC4529071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Florez L, Herrmann C, Cramer JM, Hauser CP, Koynov K, Landfester K, Crespy D, Mailander V. How shape influences uptake: interactions of anisotropic polymer nanoparticles and human mesenchymal stem cells. Small. 2012;8(14):2222–30. doi: 10.1002/smll.201102002. Epub 2012/04/25. PubMed PMID: 22528663. [DOI] [PubMed] [Google Scholar]
- 50.Barua S, Yoo JW, Kolhar P, Wakankar A, Gokarn YR, Mitragotri S. Particle shape enhances specificity of antibody-displaying nanoparticles. Proc Natl Acad Sci U S A. 2013;110(9):3270–5. doi: 10.1073/pnas.1216893110. Epub 2013/02/13. PubMed PMID: 23401509; PMCID: PMC3587278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sharma G, Valenta DT, Altman Y, Harvey S, Xie H, Mitragotri S, Smith JW. Polymer particle shape independently influences binding and internalization by macrophages. J Control Release. 2010;147(3):408–12. doi: 10.1016/j.jconrel.2010.07.116. Epub 2010/08/10. PubMed PMID: 20691741; PMCID: PMC2975856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Perica K, Tu A, Richter A, Bieler JG, Edidin M, Schneck JP. Magnetic field-induced T cell receptor clustering by nanoparticles enhances T cell activation and stimulates antitumor activity. ACS Nano. 2014;8(3):2252–60. doi: 10.1021/nn405520d. Epub 2014/02/26. PubMed PMID: 24564881; PMCID: PMC4004316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Mannix RJ, Kumar S, Cassiola F, Montoya-Zavala M, Feinstein E, Prentiss M, Ingber DE. Nanomagnetic actuation of receptor-mediated signal transduction. Nat Nanotechnol. 2008;3(1):36–40. doi: 10.1038/nnano.2007.418. Epub 2008/07/26. PubMed PMID: 18654448. [DOI] [PubMed] [Google Scholar]
- 54.Lee JH, Kim ES, Cho MH, Son M, Yeon SI, Shin JS, Cheon J. Artificial control of cell signaling and growth by magnetic nanoparticles. Angew Chem Int Ed Engl. 2010;49(33):5698–702. doi: 10.1002/anie.201001149. Epub 2010/07/08. PubMed PMID: 20607781. [DOI] [PubMed] [Google Scholar]