Biomaterial-Based Activation and Expansion of Tumor-Specific T Cells

Traditional tumor vaccination approaches mostly focus on activating dendritic cells (DCs) by providing them with a source of tumor antigens and/or adjuvants, which in turn activate tumor-reactive T cells. Novel biomaterial-based cancer immunotherapeutic strategies focus on directly activating and stimulating T cells through molecular cues presented on synthetic constructs with the aim of improving T cell survival, more precisely steer T cell activation and direct T cell differentiation. Synthetic artificial antigen presenting cells (aAPCs) decorated with T cell-activating ligands are being developed to induce robust tumor-specific T cell responses, essentially bypassing DCs. In this perspective, we approach these promising new technologies from an immunological angle, first by identifying the CD4+ and CD8+ T cell subtypes that are imperative for robust anti-cancer immunity and subsequently discussing the molecular cues needed to induce these cells types. We will elaborate on how biomaterials can be applied to stimulate T cells in vitro and in vivo to improve their survival, activation and function. Scaffold-based methods can also be used as delivery vehicles for adoptive transfer of T cells, including tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor expressing (CAR) T cells, while simultaneously stimulating these cells. Finally, we provide suggestions on how these insights could advance the field of biomaterial-based activation and expansion of tumor-specific T cells in the future.

[1]  M. Kneilling,et al.  Tumor-draining lymph nodes are pivotal in PD-1/PD-L1 checkpoint therapy. , 2018, JCI insight.

[2]  C. Figdor,et al.  Injectable Biomimetic Hydrogels as Tools for Efficient T Cell Expansion and Delivery , 2018, Front. Immunol..

[3]  D. Mooney,et al.  Biomaterial-assisted targeted modulation of immune cells in cancer treatment , 2018, Nature Materials.

[4]  Alan E. Rowan,et al.  Cytokine‐Functionalized Synthetic Dendritic Cells for T Cell Targeted Immunotherapies , 2018, Advanced Therapeutics.

[5]  S. Grupp,et al.  Reducing Ex Vivo Culture Improves the Antileukemic Activity of Chimeric Antigen Receptor (CAR) T Cells , 2018, Cancer Immunology Research.

[6]  L. Tang,et al.  Immunoengineering with biomaterials for enhanced cancer immunotherapy. , 2018, Wiley interdisciplinary reviews. Nanomedicine and nanobiotechnology.

[7]  Li Tang,et al.  Enhancing T cell therapy through TCR signaling-responsive nanoparticle drug delivery , 2018, Nature Biotechnology.

[8]  Zheng-Rong Lu,et al.  Drug Delivery in Cancer Therapy, Quo Vadis? , 2018, Molecular pharmaceutics.

[9]  A. Rogel,et al.  The immunobiology of CD27 and OX40 and their potential as targets for cancer immunotherapy. , 2018, Blood.

[10]  David J Mooney,et al.  Scaffolds that mimic antigen-presenting cells enable ex vivo expansion of primary T-cells , 2017, Nature Biotechnology.

[11]  C. Figdor,et al.  Synthetic immune niches for cancer immunotherapy , 2017, Nature Reviews Immunology.

[12]  Hai-Quan Mao,et al.  Biologically Inspired Design of Nanoparticle Artificial Antigen-Presenting Cells for Immunomodulation. , 2017, Nano letters.

[13]  H. Klok,et al.  The era of bioengineering: how will this affect the next generation of cancer immunotherapy? , 2017, Journal of Translational Medicine.

[14]  Reinhold Förster,et al.  Mechanisms and Dynamics of T Cell-Mediated Cytotoxicity In Vivo. , 2017, Trends in immunology.

[15]  K. Wittrup,et al.  Biopolymers codelivering engineered T cells and STING agonists can eliminate heterogeneous tumors , 2017, The Journal of clinical investigation.

[16]  S. Thomas,et al.  Progress and opportunities for enhancing the delivery and efficacy of checkpoint inhibitors for cancer immunotherapy , 2017, Advanced drug delivery reviews.

[17]  Matthias T Stephan,et al.  In situ programming of leukaemia-specific T cells using synthetic DNA nanocarriers. , 2017, Nature nanotechnology.

[18]  S. Rosenberg,et al.  LIGHT Elevation Enhances Immune Eradication of Colon Cancer Metastases. , 2017, Cancer research.

[19]  Jinchao Zhang,et al.  A simple and powerful co-delivery system based on pH-responsive metal-organic frameworks for enhanced cancer immunotherapy. , 2017, Biomaterials.

[20]  James I. Andorko,et al.  Impact of molecular weight on the intrinsic immunogenic activity of poly(beta amino esters). , 2017, Journal of biomedical materials research. Part A.

[21]  C. Figdor,et al.  Controlling T-Cell Activation with Synthetic Dendritic Cells Using the Multivalency Effect , 2017, ACS omega.

[22]  J. Tabernero,et al.  The expanding role of immunotherapy. , 2017, Cancer treatment reviews.

[23]  Tai-De Li,et al.  Improving T Cell Expansion with a Soft Touch. , 2017, Nano letters.

[24]  Chiara Bonini,et al.  T memory stem cells in health and disease , 2017, Nature Medicine.

[25]  U. V. von Andrian,et al.  The Chemokine Receptor CX3CR1 Defines Three Antigen-Experienced CD8 T Cell Subsets with Distinct Roles in Immune Surveillance and Homeostasis. , 2016, Immunity.

[26]  M. Manns,et al.  Tailored Tumor Immunogenicity Reveals Regulation of CD4 and CD8 T Cell Responses against Cancer. , 2016, Cell reports.

[27]  C. Van Waes,et al.  Established T Cell–Inflamed Tumors Rejected after Adaptive Resistance Was Reversed by Combination STING Activation and PD-1 Pathway Blockade , 2016, Cancer Immunology Research.

[28]  J. Griffiths,et al.  The immunometabolite S-2-hydroxyglutarate regulates CD8+ T-lymphocyte fate , 2016, Nature.

[29]  Ting Ni,et al.  Follicular CXCR5-expressing CD8+ T cells curtail chronic viral infection , 2016, Nature.

[30]  M. Sabatino,et al.  Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies. , 2016, Blood.

[31]  M. Richer,et al.  Regulation of effector and memory CD8(+) T cell function by inflammatory cytokines. , 2016, Cytokine.

[32]  Renier J. Brentjens,et al.  Driving CAR T-cells forward , 2016, Nature Reviews Clinical Oncology.

[33]  James I. Andorko,et al.  Intrinsic immunogenicity of rapidly-degradable polymers evolves during degradation. , 2016, Acta biomaterialia.

[34]  C. Jewell,et al.  Improving the clinical impact of biomaterials in cancer immunotherapy , 2016, Oncotarget.

[35]  D. Maloney,et al.  Accepted Article Preview : Published ahead of advance online publication , 2016 .

[36]  M. Delorenzi,et al.  Modulation of mTOR Signalling Triggers the Formation of Stem Cell-like Memory T Cells , 2016, EBioMedicine.

[37]  K. Anseth,et al.  Click Chemistry in Biomaterials, Nanomedicine, and Drug Delivery. , 2016, Biomacromolecules.

[38]  C. Figdor,et al.  Favorable overall survival in stage III melanoma patients after adjuvant dendritic cell vaccination , 2016, Oncoimmunology.

[39]  D. Maloney,et al.  Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo , 2015, Leukemia.

[40]  S. Lerouge,et al.  Chitosan thermogels for local expansion and delivery of tumor-specific T lymphocytes towards enhanced cancer immunotherapies. , 2016, Biomaterials.

[41]  J. Griffiths,et al.  S-2-hydroxyglutarate regulates CD 8 + T-lymphocyte fate , 2016 .

[42]  Michael Y. Gerner,et al.  In vivo characterization of the physicochemical properties of TLR agonist delivery that enhance vaccine immunogenicity , 2015, Nature Biotechnology.

[43]  M. Mann,et al.  Functional classification of memory CD8+ T cells by CX3CR1 expression , 2015, Nature Communications.

[44]  David J Mooney,et al.  Injectable cryogel-based whole-cell cancer vaccines , 2015, Nature Communications.

[45]  Paul A. Lyons,et al.  T cell exhaustion, costimulation and clinical outcome in autoimmunity and infection , 2015, Nature.

[46]  G. Ma,et al.  pH-Responsive Poly(D,L-lactic-co-glycolic acid) Nanoparticles with Rapid Antigen Release Behavior Promote Immune Response. , 2015, ACS nano.

[47]  J. Castle,et al.  Mutant MHC class II epitopes drive therapeutic immune responses to cancer , 2015, Nature.

[48]  S. Rosenberg,et al.  Adoptive cell transfer as personalized immunotherapy for human cancer , 2015, Science.

[49]  L. Qin,et al.  Genetic Evidence That Intratumoral T-cell Proliferation and Activation Are Associated with Recurrence and Survival in Patients with Resected Colorectal Liver Metastases , 2015, Cancer Immunology Research.

[50]  C. Figdor,et al.  Polymer-based synthetic dendritic cells for tailoring robust and multifunctional T cell responses. , 2015, ACS chemical biology.

[51]  K. Ishii,et al.  TLR9 and STING agonists synergistically induce innate and adaptive type-II IFN , 2015, European journal of immunology.

[52]  Mark S. Sundrud,et al.  Akt inhibition enhances expansion of potent tumor-specific lymphocytes with memory cell characteristics. , 2015, Cancer research.

[53]  Youngjin Choi,et al.  Injectable, spontaneously assembling inorganic scaffolds modulate immune cells in vivo and increase vaccine efficacy , 2014, Nature Biotechnology.

[54]  S. B. Stephan,et al.  Biopolymer implants enhance the efficacy of adoptive T cell therapy , 2014, Nature Biotechnology.

[55]  John T. Chang,et al.  Molecular regulation of effector and memory T cell differentiation , 2014, Nature Immunology.

[56]  C. Figdor,et al.  Towards efficient cancer immunotherapy: advances in developing artificial antigen-presenting cells , 2014, Trends in biotechnology.

[57]  Tarek R. Fadel,et al.  A carbon nanotube-polymer composite for T-cell therapy. , 2014, Nature nanotechnology.

[58]  S. Rosenberg IL-2: The First Effective Immunotherapy for Human Cancer , 2014, The Journal of Immunology.

[59]  S. Rosenberg,et al.  Exploiting the curative potential of adoptive T‐cell therapy for cancer , 2014, Immunological reviews.

[60]  F. Sallusto,et al.  The who's who of T‐cell differentiation: Human memory T‐cell subsets , 2013, European journal of immunology.

[61]  Michael Loran Dustin,et al.  Nanoscale ligand spacing influences receptor triggering in T cells and NK cells. , 2013, Nano letters.

[62]  Michael Loran Dustin,et al.  T Cell Activation is Determined by the Number of Presented Antigens , 2013, Nano letters.

[63]  A. Scheffold,et al.  Fine Tuning and Efficient T Cell Activation with Stimulatory aCD3 Nanoarrays , 2013, Nano letters.

[64]  C. Figdor,et al.  Therapeutic nanoworms: towards novel synthetic dendritic cells for immunotherapy , 2013 .

[65]  J. Banchereau,et al.  Dendritic-cell-based therapeutic cancer vaccines. , 2013, Immunity.

[66]  Darrell J Irvine,et al.  In vivo targeting of adoptively transferred T-cells with antibody- and cytokine-conjugated liposomes. , 2013, Journal of controlled release : official journal of the Controlled Release Society.

[67]  C. Figdor,et al.  The nature of activatory and tolerogenic dendritic cell-derived signal II , 2013, Front. Immunol..

[68]  S. Bicciato,et al.  IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. , 2013, Blood.

[69]  P. Fisher,et al.  Therapeutic cancer vaccines: past, present, and future. , 2013, Advances in cancer research.

[70]  L. Galluzzi,et al.  Tumor-Infiltrating Regulatory T Cells: Phenotype, Role, Mechanism of Expansion In Situ and Clinical Significance , 2013, Cancer Microenvironment.

[71]  Molly M. Stevens,et al.  Designing Regenerative Biomaterial Therapies for the Clinic , 2012, Science Translational Medicine.

[72]  J. O’Shea,et al.  Helper T cell diversity and plasticity. , 2012, Current opinion in immunology.

[73]  Wayne W. Hancock,et al.  Substrate Rigidity Regulates Human T Cell Activation and Proliferation , 2012, The Journal of Immunology.

[74]  S. Hvilsted Facile design of biomaterials by ‘click’ chemistry , 2012 .

[75]  Steven A. Rosenberg,et al.  Adoptive immunotherapy for cancer: harnessing the T cell response , 2012, Nature Reviews Immunology.

[76]  Karolina Palucka,et al.  Cancer immunotherapy via dendritic cells , 2012, Nature Reviews Cancer.

[77]  C. Sautès-Fridman,et al.  The immune contexture in human tumours: impact on clinical outcome , 2012, Nature Reviews Cancer.

[78]  D. Schendel,et al.  Third generation dendritic cell vaccines for tumor immunotherapy. , 2012, European journal of cell biology.

[79]  S. Kern,et al.  Th17 cells are long lived and retain a stem cell-like molecular signature. , 2011, Immunity.

[80]  G. Kroemer,et al.  Prognostic and predictive impact of intra- and peritumoral immune infiltrates. , 2011, Cancer research.

[81]  B. Alarcón,et al.  The immunological synapse: a cause or consequence of T‐cell receptor triggering? , 2011, Immunology.

[82]  F. Marincola,et al.  A human memory T-cell subset with stem cell-like properties , 2011, Nature Medicine.

[83]  S. Rosenberg,et al.  Determinants of Successful CD8+ T-Cell Adoptive Immunotherapy for Large Established Tumors in Mice , 2011, Clinical Cancer Research.

[84]  E John Wherry,et al.  T cell exhaustion , 2011 .

[85]  S. Steinberg,et al.  Durable Complete Responses in Heavily Pretreated Patients with Metastatic Melanoma Using T-Cell Transfer Immunotherapy , 2011, Clinical Cancer Research.

[86]  M. Glennie,et al.  Control of Established Melanoma by CD27 Stimulation Is Associated With Enhanced Effector Function and Persistence, and Reduced PD-1 Expression of Tumor Infiltrating CD8+ T Cells , 2010, Journal of immunotherapy.

[87]  P. J. Sanchez,et al.  Comparison of OX40 Ligand and CD70 in the Promotion of CD4+ T Cell Responses , 2010, The Journal of Immunology.

[88]  Soong Ho Um,et al.  Therapeutic cell engineering using surface-conjugated synthetic nanoparticles , 2010, Nature Medicine.

[89]  J. Curtsinger,et al.  Inflammatory cytokines as a third signal for T cell activation. , 2010, Current opinion in immunology.

[90]  Mi-Hua Tao,et al.  Cutting Edge: Mechanical Forces Acting on T Cells Immobilized via the TCR Complex Can Trigger TCR Signaling , 2010, The Journal of Immunology.

[91]  W. Paul,et al.  Differentiation of effector CD4 T cell populations (*). , 2010, Annual review of immunology.

[92]  R. Blasberg,et al.  Tumor-reactive CD4+ T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts , 2010, The Journal of experimental medicine.

[93]  P. Muranski,et al.  Naive tumor-specific CD4+ T cells differentiated in vivo eradicate established melanoma , 2010, The Journal of experimental medicine.

[94]  P. Hwu,et al.  T helper 17 cells promote cytotoxic T cell activation in tumor immunity. , 2009, Immunity.

[95]  S. Hollister Scaffold Design and Manufacturing: From Concept to Clinic , 2009, Advanced materials.

[96]  P. Muranski,et al.  Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells , 2009, Nature Medicine.

[97]  W. Paul,et al.  IL-1 acts directly on CD4 T cells to enhance their antigen-driven expansion and differentiation , 2009, Proceedings of the National Academy of Sciences.

[98]  P. Muranski,et al.  Adoptive immunotherapy of cancer using CD4(+) T cells. , 2009, Current opinion in immunology.

[99]  Samir Mitragotri,et al.  Physical approaches to biomaterial design. , 2009, Nature materials.

[100]  S. Albani,et al.  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 , 2008, Haematologica.

[101]  P. Muranski,et al.  Tumor-specific Th17-polarized cells eradicate large established melanoma. , 2008, Blood.

[102]  M. Roederer,et al.  T-cell quality in memory and protection: implications for vaccine design , 2008, Nature Reviews Immunology.

[103]  L. Sherman,et al.  Tumor-Specific CD4+ T Cells Render the Tumor Environment Permissive for Infiltration by Low-Avidity CD8+ T Cells1 , 2008, The Journal of Immunology.

[104]  C. June,et al.  4-1BB Is Superior to CD28 Costimulation for Generating CD8+ Cytotoxic Lymphocytes for Adoptive Immunotherapy1 , 2007, The Journal of Immunology.

[105]  M. Del Vecchio,et al.  Interleukin-12: Biological Properties and Clinical Application , 2007, Clinical Cancer Research.

[106]  R. Steinman,et al.  A subset of dendritic cells induces CD4+ T cells to produce IFN-γ by an IL-12–independent but CD70-dependent mechanism in vivo , 2007, The Journal of experimental medicine.

[107]  Michael Y. Gerner,et al.  Signals required for programming effector and memory development by CD8+ T cells , 2006, Immunological reviews.

[108]  S. Rosenberg,et al.  Telomere Length of Transferred Lymphocytes Correlates with In Vivo Persistence and Tumor Regression in Melanoma Patients Receiving Cell Transfer Therapy1 , 2005, The Journal of Immunology.

[109]  D. Holdstock Past, present--and future? , 2005, Medicine, conflict, and survival.

[110]  R. Langer,et al.  Smart Biomaterials , 2004, Science.

[111]  P. Doherty,et al.  Hierarchies in Cytokine Expression Profiles for Acute and Resolving Influenza Virus-Specific CD8+ T Cell Responses: Correlation of Cytokine Profile and TCR Avidity1 , 2004, The Journal of Immunology.

[112]  J. Borst,et al.  CD27 Promotes Survival of Activated T Cells and Complements CD28 in Generation and Establishment of the Effector T Cell Pool , 2003, The Journal of experimental medicine.

[113]  M. Glennie,et al.  Expression and costimulatory effects of the TNF receptor superfamily members CD134 (OX40) and CD137 (4‐1BB), and their role in the generation of anti‐tumor immune responses , 2002, European journal of immunology.

[114]  I. Frazer,et al.  Antigen-specific CD4+ T-cell help is required to activate a memory CD8+ T cell to a fully functional tumor killer cell. , 2002, Cancer research.

[115]  G. Zhu,et al.  Modulation of T-cell-mediated immunity in tumor and graft-versus-host disease models through the LIGHT co-stimulatory pathway , 2000, Nature Medicine.

[116]  A. Ohta,et al.  The critical role of Th1-dominant immunity in tumor immunology , 2000, Cancer Chemotherapy and Pharmacology.

[117]  R. Steinman,et al.  Dendritic cells and the control of immunity , 1998, Nature.

[118]  R. Germain,et al.  Single Cell Analysis Reveals Regulated Hierarchical T Cell Antigen Receptor Signaling Thresholds and Intraclonal Heterogeneity for Individual Cytokine Responses of CD4+ T Cells , 1997, The Journal of experimental medicine.

[119]  P. Lipsky,et al.  Accessory cell independent proliferation of human T4 cells stimulated by immobilized monoclonal antibodies to CD3. , 1987, Journal of immunology.