Engineering Solutions for Mitigation of Chimeric Antigen Receptor T-Cell Dysfunction
Abstract
:1. Introduction
1.1. The Molecular Regulators of Exhaustion
1.2. Defining CAR-T-Cell Exhaustion and Dysfunction
2. Engineering Solutions
2.1. Chimeric Antigen Receptor and Vector Design
2.1.1. Antigen Binding Domain
2.1.2. Spacer and Transmembrane
2.1.3. Signalling Domain
2.1.4. Regulating CAR Expression and Function
2.2. Optimising Manufacturing Steps
2.2.1. Cell Product Selection Steps
2.2.2. Activation Methods
2.2.3. Ex Vivo Expansion Milieu
2.2.4. Drugs to Regulate T-Cell Development
2.3. Transcriptional Reprogramming to Mitigate or Exploit Exhaustion
2.4. Using Boolean Logic and Switch Systems to Limit Stimulation and Mitigate Exhaustion
Beyond Basic Logic; Sequential Logic Gates and Molecular Switches
2.5. Creating A Pro-Inflammatory Host/Tumour Microenvironment
2.5.1. Administering Cytokines
2.5.2. Engineering Cytokine Signalling
2.5.3. Targeting Immunosuppressive Mechanisms
3. Conclusions
Funding
Conflicts of Interest
References
- Morgan, R.A.; Dudley, M.E.; Wunderlich, J.R.; Hughes, M.S.; Yang, J.C.; Sherry, R.M.; Royal, R.E.; Topalian, S.L.; Kammula, U.S.; Restifo, N.P.; et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 2006, 314, 126–129. [Google Scholar] [CrossRef] [Green Version]
- Gross, G.; Waks, T.; Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl. Acad. Sci. USA 1989, 86, 10024–10028. [Google Scholar] [CrossRef] [Green Version]
- Gross, G.; Gorochov, G.; Waks, T.; Eshhar, Z. Generation of effector T cells expressing chimeric T cell receptor with antibody type-specificity. Transplant. Proc. 1989, 21, 127–130. [Google Scholar]
- Kuwana, Y.; Asakura, Y.; Utsunomiya, N.; Nakanishi, M.; Arata, Y.; Itoh, S.; Nagase, F.; Kurosawa, Y. Expression of chimeric receptor composed of immunoglobulin-derived V resions and T-cell receptor-derived C regions. Biochem. Biophys. Res. Commun. 1987, 149, 960–968. [Google Scholar] [CrossRef]
- Sadelain, M.; Rivière, I.; Riddell, S. Therapeutic T cell engineering. Nature 2017, 545, 423–431. [Google Scholar] [CrossRef] [Green Version]
- Maude, S.L.; Frey, N.; Shaw, P.A.; Aplenc, R.; Barrett, D.M.; Bunin, N.J.; Chew, A.; Gonzalez, V.E.; Zheng, Z.; Lacey, S.F.; et al. Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia. N. Engl. J. Med. 2014, 371, 1507–1517. [Google Scholar] [CrossRef] [Green Version]
- Titov, A.; Petukhov, A.; Staliarova, A.; Motorin, D.; Bulatov, E.; Shuvalov, O.; Soond, S.M.; Piacentini, M.; Melino, G.; Zaritskey, A.; et al. The biological basis and clinical symptoms of CAR-T therapy-associated toxicites. Cell Death Dis. 2018, 9, 897. [Google Scholar] [CrossRef]
- Majzner, R.G.; Mackall, C.L. Clinical lessons learned from the first leg of the CAR T cell journey. Nat. Med. 2019, 25, 1341–1355. [Google Scholar] [CrossRef] [PubMed]
- Martinez, M.; Moon, E.K. CAR T Cells for Solid Tumors: New Strategies for Finding, Infiltrating, and Surviving in the Tumor Microenvironment. Front. Immunol. 2019, 10, 128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, W.-L.; Hua, Z.-C. Chimeric Antigen Receptor T-cell (CAR T) Therapy for Hematologic and Solid Malignancies: Efficacy and Safety-A Systematic Review with Meta-Analysis. Cancers 2019, 11, 47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pule, M.A.; Savoldo, B.; Myers, G.D.; Rossig, C.; Russell, H.V.; Dotti, G.; Huls, M.H.; Liu, E.; Gee, A.P.; Mei, Z.; et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: Persistence and antitumor activity in individuals with neuroblastoma. Nat. Med. 2008, 14, 1264–1270. [Google Scholar] [CrossRef] [PubMed]
- Gallimore, A.; Glithero, A.; Godkin, A.; Tissot, A.C.; Plückthun, A.; Elliott, T.; Hengartner, H.; Zinkernagel, R. Induction and Exhaustion of Lymphocytic Choriomeningitis Virus–specific Cytotoxic T Lymphocytes Visualized Using Soluble Tetrameric Major Histocompatibility Complex Class I–Peptide Complexes. J. Exp. Med. 1998, 187, 1383–1393. [Google Scholar] [CrossRef] [PubMed]
- Byers, V.S.; Sercarz, E.E. The X-Y-Z Scheme of Immunocyte Maturation. J. Exp. Med. 1968, 127, 307–325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McLane, L.M.; Abdel-Hakeem, M.S.; Wherry, E.J. CD8 T Cell Exhaustion During Chronic Viral Infection and Cancer. Annu. Rev. Immunol. 2019, 37, 457–495. [Google Scholar] [CrossRef] [Green Version]
- Shankar, P.; Russo, M.; Harnisch, B.; Patterson, M.; Skolnik, P.; Lieberman, J. Impaired function of circulating HIV-specific CD8+ T cells in chronic human immunodeficiency virus infection. Blood 2000, 96, 3094–3101. [Google Scholar] [CrossRef]
- Lechner, F.; Wong, D.K.H.; Dunbar, P.R.; Chapman, R.; Chung, R.T.; Dohrenwend, P.; Robbins, G.; Phillips, R.; Klenerman, P.; Walker, B.D. Analysis of Successful Immune Responses in Persons Infected with Hepatitis C Virus. J. Exp. Med. 2000, 191, 1499–1512. [Google Scholar] [CrossRef] [Green Version]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
- Blank, C.U.; Haining, W.N.; Held, W.; Hogan, P.G.; Kallies, A.; Lugli, E.; Lynn, R.C.; Philip, M.; Rao, A.; Restifo, N.P.; et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol. 2019, 19, 665–674. [Google Scholar] [CrossRef]
- Kallies, A.; Zehn, D.; Utzschneider, D.T. Precursor exhausted T cells: Key to successful immunotherapy? Nat. Rev. Immunol. 2019, 20, 128–136. [Google Scholar] [CrossRef]
- Beltra, J.-C.; Manne, S.; Abdel-Hakeem, M.S.; Kurachi, M.; Giles, J.R.; Chen, Z.; Casella, V.; Ngiow, S.F.; Khan, O.; Huang, Y.J.; et al. Developmental Relationships of Four Exhausted CD8+ T Cell Subsets Reveals Underlying Transcriptional and Epigenetic Landscape Control Mechanisms. Immunity 2020. [Google Scholar] [CrossRef]
- Chang, J.T.; Wherry, E.J.; Goldrath, A.W. Molecular regulation of effector and memory T cell differentiation. Nat. Immunol. 2014, 15, 1104–1115. [Google Scholar] [CrossRef] [PubMed]
- Zajac, A.J.; Blattman, J.N.; Murali-Krishna, K.; Sourdive, D.J.D.; Suresh, M.; Altman, J.D.; Ahmed, R. Viral Immune Evasion Due to Persistence of Activated T Cells Without Effector Function. J. Exp. Med. 1998, 188, 2205–2213. [Google Scholar] [CrossRef]
- Moskophidis, D.; Lechner, F.; Pircher, H.; Zinkernagel, R.M. Virus persistence in acutely infected immunocompetent mice by exhaustion of antiviral cytotoxic effector T cells. Nature 1993, 362, 758–761. [Google Scholar] [CrossRef] [PubMed]
- Wherry, E.J.; Ha, S.-J.; Kaech, S.M.; Haining, W.N.; Sarkar, S.; Kalia, V.; Subramaniam, S.; Blattman, J.N.; Barber, D.L.; Ahmed, R. Molecular Signature of CD8+ T Cell Exhaustion during Chronic Viral Infection. Immunity 2007, 27, 670–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Z.; Ji, Z.; Ngiow, S.F.; Manne, S.; Cai, Z.; Huang, A.C.; Johnson, J.; Staupe, R.P.; Bengsch, B.; Xu, C.; et al. TCF-1-Centered Transcriptional Network Drives an Effector versus Exhausted CD8 T Cell-Fate Decision. Immunity 2019, 51, 840–855.e5. [Google Scholar] [CrossRef]
- Utzschneider, D.T.; Legat, A.; Marraco, S.A.F.; Carrié, L.; Luescher, I.; Speiser, D.E.; Zehn, D. T cells maintain an exhausted phenotype after antigen withdrawal and population reexpansion. Nat. Immunol. 2013, 14, 603–610. [Google Scholar] [CrossRef]
- Miller, B.C.; Sen, D.R.; Abosy, R.A.; Bi, K.; Virkud, Y.V.; LaFleur, M.W.; Yates, K.B.; Lako, A.; Felt, K.; Naik, G.S.; et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 2019, 20, 326–336. [Google Scholar] [CrossRef]
- Verma, V.; Shrimali, R.K.; Ahmad, S.; Dai, W.; Wang, H.; Lu, S.; Nandre, R.; Gaur, P.; Lopez, J.; Sade-Feldman, M.; et al. PD-1 blockade in subprimed CD8 cells induces dysfunctional PD-1+CD38hi cells and anti-PD-1 resistance. Nat. Immunol. 2019, 20, 1231–1243. [Google Scholar] [CrossRef]
- Schietinger, A.; Philip, M.; Krisnawan, V.E.; Chiu, E.Y.; Delrow, J.J.; Basom, R.S.; Lauer, P.; Brockstedt, D.G.; Knoblaugh, S.E.; Hämmerling, G.J.; et al. Tumor-Specific T Cell Dysfunction Is a Dynamic Antigen-Driven Differentiation Program Initiated Early during Tumorigenesis. Immunity 2016, 45, 389–401. [Google Scholar] [CrossRef] [Green Version]
- Pauken, K.E.; Sammons, M.A.; Odorizzi, P.M.; Manne, S.; Godec, J.; Khan, O.; Drake, A.M.; Chen, Z.; Sen, D.R.; Kurachi, M.; et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 2016, 354, 1160–1165. [Google Scholar] [CrossRef] [Green Version]
- Khan, O.; Giles, J.R.; McDonald, S.; Manne, S.; Ngiow, S.F.; Patel, K.P.; Werner, M.T.; Huang, A.C.; Alexander, K.A.; Wu, J.E.; et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 2019, 571, 211–218. [Google Scholar] [CrossRef] [PubMed]
- Yao, C.; Sun, H.-W.; Lacey, N.E.; Ji, Y.; Moseman, E.A.; Shih, H.-Y.; Heuston, E.F.; Kirby, M.; Anderson, S.; Cheng, J.; et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection. Nat. Immunol. 2019, 20, 890–901. [Google Scholar] [CrossRef] [PubMed]
- Seo, H.; Chen, J.; González-Avalos, E.; Samaniego-Castruita, D.; Das, A.; Wang, Y.H.; López-Moyado, I.F.; Georges, R.O.; Zhang, W.; Onodera, A.; et al. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8+ T cell exhaustion. Proc. Natl. Acad. Sci. USA 2019, 116, 12410–12415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alfei, F.; Kanev, K.; Hofmann, M.; Wu, M.; Ghoneim, H.E.; Roelli, P.; Utzschneider, D.T.; von Hoesslin, M.; Cullen, J.G.; Fan, Y.; et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 2019, 571, 265–269. [Google Scholar] [CrossRef] [PubMed]
- Scott, A.C.; Dündar, F.; Zumbo, P.; Chandran, S.S.; Klebanoff, C.A.; Shakiba, M.; Trivedi, P.; Menocal, L.; Appleby, H.; Camara, S.; et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 2019, 571, 1–5. [Google Scholar] [CrossRef]
- Mann, T.H.; Kaech, S.M. Tick-TOX, it’s time for T cell exhaustion. Nat. Immunol. 2019, 20, 1092–1094. [Google Scholar] [CrossRef]
- Zhang, Z.; Liu, S.; Zhang, B.; Qiao, L.; Zhang, Y.; Zhang, Y. T Cell Dysfunction and Exhaustion in Cancer. Front. Cell Dev. Biol. 2020, 8, 17. [Google Scholar] [CrossRef] [Green Version]
- Aliahmad, P.; O’Flaherty, E.; Han, P.; Goularte, O.D.; Wilkinson, B.; Satake, M.; Molkentin, J.D.; Kaye, J. TOX Provides a Link Between Calcineurin Activation and CD8 Lineage Commitment. J. Exp. Med. 2004, 199, 1089–1099. [Google Scholar] [CrossRef]
- Chen, J.; López-Moyado, I.F.; Seo, H.; Lio, C.-W.J.; Hempleman, L.J.; Sekiya, T.; Yoshimura, A.; Scott-Browne, J.P.; Rao, A. NR4A transcription factors limit CAR T cell function in solid tumours. Nature 2019, 567, 530–534. [Google Scholar] [CrossRef]
- Li, F.; Zhang, Y. Targeting NR4As, a new strategy to fine-tune CAR-T cells against solid tumors. Signal Transduct. Target. Ther. 2019, 4, 7. [Google Scholar] [CrossRef]
- Hajaj, E.; Eisenberg, G.; Klein, S.; Frankenburg, S.; Merims, S.; David, I.B.; Eisenhaure, T.; Henrickson, S.E.; Villani, A.C.; Hacohen, N.; et al. SLAMF6 deficiency augments tumor killing and skews towards an effector phenotype revealing it as a novel T cell checkpoint. Elife 2020, 9, e52539. [Google Scholar] [CrossRef] [PubMed]
- Lynn, R.C.; Weber, E.W.; Sotillo, E.; Gennert, D.; Xu, P.; Good, Z.; Anbunathan, H.; Lattin, J.; Jones, R.; Tieu, V.; et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 2019, 576, 293–300. [Google Scholar] [CrossRef] [PubMed]
- Long, A.H.; Haso, W.M.; Shern, J.F.; Wanhainen, K.M.; Murgai, M.; Ingaramo, M.; Smith, J.P.; Walker, A.J.; Kohler, M.E.; Venkateshwara, V.R.; et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. 2015, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Frigault, M.J.; Lee, J.; Basil, M.C.; Carpenito, C.; Motohashi, S.; Scholler, J.; Kawalekar, O.U.; Guedan, S.; McGettigan, S.E.; Posey, A.D.; et al. Identification of chimeric antigen receptors that mediate constitutive or inducible proliferation of T cells. Cancer Immunol. Res. 2015, 3, 356–367. [Google Scholar] [CrossRef] [Green Version]
- Fisher, J.; Sharma, R.; Don, D.; Barisa, M.; Hurtado, M.; Abramowski, P.; Porter, L.; Day, W.; Borea, R.; Inglott, S.; et al. Engineering γδT cells limits tonic signaling associated with chimeric antigen receptors. Sci. Signal. 2019, 12, eaax1872. [Google Scholar] [CrossRef]
- Watanabe, N.; Bajgain, P.; Sukumaran, S.; Ansari, S.; Heslop, H.E.; Rooney, C.M.; Brenner, M.K.; Leen, A.M.; Vera, J.F. Fine-tuning the CAR spacer improves T-cell potency. OncoImmunology 2016, 5, e1253656. [Google Scholar] [CrossRef] [Green Version]
- Hombach, A.; Hombach, A.A.; Abken, H. Adoptive immunotherapy with genetically engineered T cells: Modification of the IgG1 Fc “spacer” domain in the extracellular moiety of chimeric antigen receptors avoids “off-target” activation and unintended initiation of an innate immune response. Gene Ther. 2010, 17, 1206–1213. [Google Scholar] [CrossRef] [Green Version]
- Hudecek, M.; Sommermeyer, D.; Kosasih, P.L.; Silva-Benedict, A.; Liu, L.; Rader, C.; Jensen, M.C.; Riddell, S.R. The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol. Res. 2015, 3, 125–135. [Google Scholar] [CrossRef] [Green Version]
- Chmielewski, M.; Hombach, A.; Heuser, C.; Adams, G.P.; Abken, H. T Cell Activation by Antibody-Like Immunoreceptors: Increase in Affinity of the Single-Chain Fragment Domain above Threshold Does Not Increase T Cell Activation against Antigen-Positive Target Cells but Decreases Selectivity. J. Immunol. 2004, 173, 7647–7653. [Google Scholar] [CrossRef]
- Liu, X.; Jiang, S.; Fang, C.; Yang, S.; Olalere, D.; Pequignot, E.C.; Cogdill, A.P.; Li, N.; Ramones, M.; Granda, B.; et al. Affinity-Tuned ErbB2 or EGFR Chimeric Antigen Receptor T Cells Exhibit an Increased Therapeutic Index against Tumors in Mice. Cancer Res. 2015, 75, 3596–3607. [Google Scholar] [CrossRef] [Green Version]
- Caruso, H.G.; Hurton, L.V.; Najjar, A.; Rushworth, D.; Ang, S.; Olivares, S.; Mi, T.; Switzer, K.; Singh, H.; Huls, H.; et al. Tuning Sensitivity of CAR to EGFR Density Limits Recognition of Normal Tissue While Maintaining Potent Antitumor Activity. Cancer Res. 2015, 75, 3505–3518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, S.; Shevlin, E.; Vedvyas, Y.; Zaman, M.; Park, S.; Hsu, Y.-M.S.; Min, I.M.; Jin, M.M. Micromolar affinity CAR T cells to ICAM-1 achieves rapid tumor elimination while avoiding systemic toxicity. Sci. Rep. (UK) 2017, 7, 14366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hudecek, M.; Lupo-Stanghellini, M.T.; Kosasih, P.L.; Sommermeyer, D.; Jensen, M.C.; Rader, C.; Riddell, S.R. Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin. Cancer Res. 2013, 19, 3153–3164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ghorashian, S.; Kramer, A.M.; Onuoha, S.; Wright, G.; Bartram, J.; Richardson, R.; Albon, S.J.; Casanovas-Company, J.; Castro, F.; Popova, B.; et al. Enhanced CAR T cell expansion and prolonged persistence in pediatric patients with ALL treated with a low-affinity CD19 CAR. Nat. Med. 2019, 25, 1408–1414. [Google Scholar] [CrossRef]
- Jonnalagadda, M.; Mardiros, A.; Urak, R.; Wang, X.; Hoffman, L.J.; Bernanke, A.; Chang, W.-C.; Bretzlaff, W.; Starr, R.; Priceman, S.; et al. Chimeric antigen receptors with mutated IgG4 Fc spacer avoid fc receptor binding and improve T cell persistence and antitumor efficacy. Mol. Ther. 2015, 23, 757–768. [Google Scholar] [CrossRef] [Green Version]
- Almåsbak, H.; Walseng, E.; Kristian, A.; Myhre, M.R.; Suso, E.M.; Munthe, L.A.; Andersen, J.T.; Wang, M.Y.; Kvalheim, G.; Gaudernack, G.; et al. Inclusion of an IgG1-Fc spacer abrogates efficacy of CD19 CAR T cells in a xenograft mouse model. Gene Ther. 2015, 22, 391–403. [Google Scholar] [CrossRef]
- Majzner, R.G.; Rietberg, S.P.; Sotillo, E.; Dong, R.; Vachharajani, V.T.; Labanieh, L.; Myklebust, J.H.; Kadapakkam, M.; Weber, E.W.; Tousley, A.M.; et al. Tuning the Antigen Density Requirement for CAR T-cell Activity. Cancer Discov. 2020, 10, 702–723. [Google Scholar] [CrossRef] [Green Version]
- Maher, J.; Brentjens, R.J.; Gunset, G.; Rivière, I.; Sadelain, M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRζ /CD28 receptor. Nat. Biotechnol. 2002, 20, 70–75. [Google Scholar] [CrossRef]
- Brentjens, R.J.; Davila, M.L.; Rivière, I.; Park, J.; Wang, X.; Cowell, L.G.; Bartido, S.; Stefanski, J.; Taylor, C.; Olszewska, M.; et al. CD19-Targeted T Cells Rapidly Induce Molecular Remissions in Adults with Chemotherapy-Refractory Acute Lymphoblastic Leukemia. Sci. Transl. Med. 2013, 5, 177ra38. [Google Scholar] [CrossRef] [Green Version]
- Porter, D.L.; Hwang, W.-T.; Frey, N.V.; Lacey, S.F.; Shaw, P.A.; Loren, A.W.; Bagg, A.; Marcucci, K.T.; Shen, A.; Gonzalez, V.; et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci. Transl. Med. 2015, 7, 303ra139. [Google Scholar] [CrossRef] [Green Version]
- Kawalekar, O.U.; O’Connor, R.S.; Fraietta, J.A.; Guo, L.; McGettigan, S.E.; Posey, A.D.; Patel, P.R.; Guedan, S.; Scholler, J.; Keith, B.; et al. Distinct Signaling of Coreceptors Regulates Specific Metabolism Pathways and Impacts Memory Development in CAR T Cells. Immunity 2016, 44, 380–390. [Google Scholar] [CrossRef] [PubMed]
- Salter, A.I.; Ivey, R.G.; Kennedy, J.J.; Voillet, V.; Rajan, A.; Alderman, E.J.; Voytovich, U.J.; Lin, C.; Sommermeyer, D.; Liu, L.; et al. Phosphoproteomic analysis of chimeric antigen receptor signaling reveals kinetic and quantitative differences that affect cell function. Sci. Signal. 2018, 11, eaat6753. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Finney, H.M.; Akbar, A.N.; Lawson, A.D.G. Activation of Resting Human Primary T Cells with Chimeric Receptors: Costimulation from CD28, Inducible Costimulator, CD134, and CD137 in Series with Signals from the TCRζ Chain. J. Immunol. 2003, 172, 104–113. [Google Scholar] [CrossRef] [PubMed]
- Guedan, S.; Chen, X.; Madar, A.; Carpenito, C.; McGettigan, S.E.; Frigault, M.J.; Lee, J.; Posey, A.D.; Scholler, J.; Scholler, N.; et al. ICOS-based chimeric antigen receptors program bipolar TH17/TH1 cells. Blood 2014, 124, 1070–1080. [Google Scholar] [CrossRef] [Green Version]
- Guedan, S.; Posey, A.D.; Shaw, C.; Wing, A.; Da, T.; Patel, P.R.; McGettigan, S.E.; Casado-Medrano, V.; Kawalekar, O.U.; Uribe-Herranz, M.; et al. Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation. JCI Insight 2018, 3, e96976. [Google Scholar] [CrossRef] [Green Version]
- Pule, M.A.; Straathof, K.C.; Dotti, G.; Heslop, H.E.; Rooney, C.M.; Brenner, M.K. A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells. Mol. Ther. 2005, 12, 933–941. [Google Scholar] [CrossRef]
- Imai, C.; Mihara, K.; Andreansky, M.; Nicholson, I.C.; Pui, C.-H.; Geiger, T.L.; Campana, D. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia 2004, 18, 676–684. [Google Scholar] [CrossRef] [Green Version]
- Song, D.-G.; Ye, Q.; Poussin, M.; Harms, G.M.; Figini, M.; Powell, D.J. CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivo. Blood 2012, 119, 696–706. [Google Scholar] [CrossRef]
- Eyquem, J.; Mansilla-Soto, J.; Giavridis, T.; van der Stegen, S.J.C.; Hamieh, M.; Cunanan, K.M.; Odak, A.; Gonen, M.; Sadelain, M. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 2017, 543, 113–117. [Google Scholar] [CrossRef] [Green Version]
- Feucht, J.; Sun, J.; Eyquem, J.; Ho, Y.-J.; Zhao, Z.; Leibold, J.; Dobrin, A.; Cabriolu, A.; Hamieh, M.; Sadelain, M. Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency. Nat. Med. 2019, 25, 82–88. [Google Scholar] [CrossRef]
- Walker, A.J.; Majzner, R.G.; Zhang, L.; Wanhainen, K.; Long, A.H.; Nguyen, S.M.; Lopomo, P.; Vigny, M.; Fry, T.J.; Orentas, R.J.; et al. Tumor Antigen and Receptor Densities Regulate Efficacy of a Chimeric Antigen Receptor Targeting Anaplastic Lymphoma Kinase. Mol. Ther. 2017, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Smith, E.L.; Staehr, M.; Masakayan, R.; Tatake, I.J.; Purdon, T.J.; Wang, X.; Wang, P.; Liu, H.; Xu, Y.; Garrett-Thomson, S.C.; et al. Development and Evaluation of an Optimal Human Single-Chain Variable Fragment-Derived BCMA-Targeted CAR T Cell Vector. Mol. Ther. J. Am. Soc. Gene Ther. 2018, 26, 1447–1456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alonso-Camino, V.; Sánchez-Martín, D.; Compte, M.; Nuñez-Prado, N.; Diaz, R.M.; Vile, R.; Alvarez-Vallina, L. CARbodies: Human Antibodies Against Cell Surface Tumor Antigens Selected From Repertoires Displayed on T Cell Chimeric Antigen Receptors. Mol. Ther. Nucleic Acids 2013, 2, e93. [Google Scholar] [CrossRef] [PubMed]
- Kuznetsova, M.; Lopatnikova, J.; Shevchenko, J.; Silkov, A.; Maksyutov, A.; Sennikov, S. Cytotoxic Activity and Memory T Cell Subset Distribution of in vitro-Stimulated CD8+ T Cells Specific for HER2/neu Epitopes. Front. Immunol. 2019, 10, 1017. [Google Scholar] [CrossRef]
- Liadi, I.; Singh, H.; Romain, G.; Rey-Villamizar, N.; Merouane, A.; Adolacion, J.R.T.; Kebriaei, P.; Huls, H.; Qiu, P.; Roysam, B.; et al. Individual Motile CD4(+) T Cells Can Participate in Efficient Multikilling through Conjugation to Multiple Tumor Cells. Cancer Immunol. Res. 2015, 3, 473–482. [Google Scholar] [CrossRef] [Green Version]
- Turtle, C.J.; Hanafi, L.-A.; Berger, C.; Gooley, T.A.; Cherian, S.; Hudecek, M.; Sommermeyer, D.; Melville, K.; Pender, B.; Budiarto, T.M.; et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Investig. 2016, 126, 2123–2138. [Google Scholar] [CrossRef] [Green Version]
- Riddell, S.R.; Sommermeyer, D.; Berger, C.; Liu, L.S.; Balakrishnan, A.; Salter, A.; Hudecek, M.; Maloney, D.G.; Turtle, C.J. Adoptive therapy with chimeric antigen receptor-modified T cells of defined subset composition. Cancer J. (Sudbury Mass.) 2014, 20, 141–144. [Google Scholar] [CrossRef] [Green Version]
- Hinrichs, C.S.; Gattinoni, L.; Restifo, N.P. Programming CD8+ T cells for effective immunotherapy. Curr. Opin. Immunol. 2006, 18, 363–370. [Google Scholar] [CrossRef] [Green Version]
- Hinrichs, C.S.; Borman, Z.A.; Cassard, L.; Gattinoni, L.; Spolski, R.; Yu, Z.; Sanchez-Perez, L.; Muranski, P.; Kern, S.J.; Logun, C.; et al. Adoptively transferred effector cells derived from naive rather than central memory CD8+ T cells mediate superior antitumor immunity. Proc. Natl. Acad. Sci. USA 2009, 106, 17469–17474. [Google Scholar] [CrossRef] [Green Version]
- Gattinoni, L.; Klebanoff, C.A.; Palmer, D.C.; Wrzesinski, C.; Kerstann, K.; Yu, Z.; Finkelstein, S.E.; Theoret, M.R.; Rosenberg, S.A.; Restifo, N.P. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J. Clin. Investig. 2005, 115, 1616–1626. [Google Scholar] [CrossRef]
- Gattinoni, L.; Lugli, E.; Ji, Y.; Pos, Z.; Paulos, C.M.; Quigley, M.F.; Almeida, J.R.; Gostick, E.; Yu, Z.; Carpenito, C.; et al. A human memory T cell subset with stem cell-like properties. Nat. Med. 2011, 17, 1290–1297. [Google Scholar] [CrossRef] [PubMed]
- Hinrichs, C.S.; Borman, Z.A.; Gattinoni, L.; Yu, Z.; Burns, W.R.; Huang, J.; Klebanoff, C.A.; Johnson, L.A.; Kerkar, S.P.; Yang, S.; et al. Human effector CD8+ T cells derived from naive rather than memory subsets possess superior traits for adoptive immunotherapy. Blood 2011, 117, 808–814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Berger, C.; Jensen, M.C.; Lansdorp, P.M.; Gough, M.; Elliott, C.; Riddell, S.R. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J. Clin. Investig. 2008, 118, 294–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, X.; Naranjo, A.; Brown, C.E.; Bautista, C.; Wong, C.W.; Chang, W.-C.; Aguilar, B.; Ostberg, J.R.; Riddell, S.R.; Forman, S.J.; et al. Phenotypic and functional attributes of lentivirus-modified CD19-specific human CD8+ central memory T cells manufactured at clinical scale. J. Immunother. (Hagerstown MD 1997) 2012, 35, 689–701. [Google Scholar] [CrossRef] [Green Version]
- Conlon, K.C.; Potter, E.L.; Pittaluga, S.; Lee, C.-C.R.; Miljkovic, M.D.; Fleisher, T.A.; Dubois, S.; Bryant, B.R.; Petrus, M.; Perera, L.P.; et al. IL15 by Continuous Intravenous Infusion to Adult Patients with Solid Tumors in a Phase I Trial Induced Dramatic NK-Cell Subset Expansion. Clin. Cancer Res. 2019, 25, 4945–4954. [Google Scholar] [CrossRef]
- Klebanoff, C.A.; Scott, C.D.; Leonardi, A.J.; Yamamoto, T.N.; Cruz, A.C.; Ouyang, C.; Ramaswamy, M.; Roychoudhuri, R.; Ji, Y.; Eil, R.L.; et al. Memory T cell-driven differentiation of naive cells impairs adoptive immunotherapy. J. Clin. Investig. 2015, 126, 318–334. [Google Scholar] [CrossRef] [Green Version]
- Stock, S.; Schmitt, M.; Sellner, L. Optimizing Manufacturing Protocols of Chimeric Antigen Receptor T Cells for Improved Anticancer Immunotherapy. Int. J. Mol. Sci. 2019, 20, 6223. [Google Scholar] [CrossRef] [Green Version]
- Casati, A.; Varghaei-Nahvi, A.; Feldman, S.A.; Assenmacher, M.; Rosenberg, S.A.; Dudley, M.E.; Scheffold, A. Clinical-scale selection and viral transduction of human naïve and central memory CD8+ T cells for adoptive cell therapy of cancer patients. Cancer Immunol. Immunother. CII 2013, 62, 1563–1573. [Google Scholar] [CrossRef]
- Barrett, D.M.; Singh, N.; Liu, X.; Jiang, S.; June, C.H.; Grupp, S.A.; Zhao, Y. Relation of clinical culture method to T-cell memory status and efficacy in xenograft models of adoptive immunotherapy. Cytotherapy 2014, 16, 619–630. [Google Scholar] [CrossRef] [Green Version]
- Gargett, T.; Brown, M.P. Different cytokine and stimulation conditions influence the expansion and immune phenotype of third-generation chimeric antigen receptor T cells specific for tumor antigen GD2. Cytotherapy 2015, 17, 487–495. [Google Scholar] [CrossRef]
- Gargett, T.; Truong, N.; Ebert, L.M.; Yu, W.; Brown, M.P. Optimization of manufacturing conditions for chimeric antigen receptor T cells to favor cells with a central memory phenotype. Cytotherapy 2019, 21, 593–602. [Google Scholar] [CrossRef] [PubMed]
- Stock, S.; Hoffmann, J.-M.; Schubert, M.-L.; Wang, L.; Wang, S.; Gong, W.; Neuber, B.; Gern, U.; Schmitt, A.; Müller-Tidow, C.; et al. Influence of Retronectin-Mediated T-Cell Activation on Expansion and Phenotype of CD19-Specific Chimeric Antigen Receptor T Cells. Hum. Gene Ther. 2018, 29, 1167–1182. [Google Scholar] [CrossRef] [PubMed]
- Kebriaei, P.; Singh, H.; Huls, M.H.; Figliola, M.J.; Bassett, R.; Olivares, S.; Jena, B.; Dawson, M.J.; Kumaresan, P.R.; Su, S.; et al. Phase I trials using Sleeping Beauty to generate CD19-specific CAR T cells. J. Clin. Investig. 2016, 126, 3363–3376. [Google Scholar] [CrossRef] [PubMed]
- Perales, M.-A.; Kebriaei, P.; Kean, L.S.; Sadelain, M. Building a Safer and Faster CAR: Seatbelts, Airbags, and CRISPR. Biol. Blood Marrow Transplant. 2018, 24, 27–31. [Google Scholar] [CrossRef] [Green Version]
- Themeli, M.; Kloss, C.C.; Ciriello, G.; Fedorov, V.D.; Perna, F.; Gonen, M.; Sadelain, M. Generation of tumor-targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy. Nat. Biotechnol. 2013, 31. [Google Scholar] [CrossRef]
- Shourian, M.; Beltra, J.-C.; Bourdin, B.; Decaluwe, H. Common gamma chain cytokines and CD8 T cells in cancer. In Seminars in Immunology; Academic Press: Cambridge, MA, USA, 2019; Volume 42, p. 101307. [Google Scholar] [CrossRef]
- Leonard, W.J.; Lin, J.-X.; O’Shea, J.J. The γc Family of Cytokines: Basic Biology to Therapeutic Ramifications. Immunity 2019, 50, 832–850. [Google Scholar] [CrossRef] [Green Version]
- Dudley, M.E.; Yang, J.C.; Sherry, R.; Hughes, M.S.; Royal, R.; Kammula, U.; Robbins, P.F.; Huang, J.; Citrin, D.E.; Leitman, S.F.; et al. Adoptive cell therapy for patients with metastatic melanoma: Evaluation of intensive myeloablative chemoradiation preparative regimens. J. Clin. Oncol. 2008, 26, 5233–5239. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Lotze, M.T.; Muul, L.M.; Leitman, S.; Chang, A.E.; Ettinghausen, S.E.; Matory, Y.L.; Skibber, J.M.; Shiloni, E.; Vetto, J.T.; et al. Observations on the Systemic Administration of Autologous Lymphokine-Activated Killer Cells and Recombinant Interleukin-2 to Patients with Metastatic Cancer. N. Engl. J. Med. 1985, 313, 1485–1492. [Google Scholar] [CrossRef]
- Alizadeh, D.; Wong, R.A.; Yang, X.; Wang, D.; Pecoraro, J.R.; Kuo, C.-F.; Aguilar, B.; Qi, Y.; Ann, D.K.; Starr, R.; et al. IL-15-mediated reduction of mTORC1 activity preserves the stem cell memory phenotype of CAR-T cells and confers superior antitumor activity. Cancer Immunol. Res. 2019, 7. [Google Scholar] [CrossRef]
- Heemskerk, B.; Liu, K.; Dudley, M.E.; Johnson, L.A.; Kaiser, A.; Downey, S.; Zheng, Z.; Shelton, T.E.; Matsuda, K.; Robbins, P.F.; et al. Adoptive cell therapy for patients with melanoma, using tumor-infiltrating lymphocytes genetically engineered to secrete interleukin-2. Hum. Gene Ther. 2008, 19, 496–510. [Google Scholar] [CrossRef]
- Bluestone, J.A. The yin and yang of interleukin-2-mediated immunotherapy. N. Engl. J. Med. 2011, 365, 2129–2131. [Google Scholar] [CrossRef] [PubMed]
- Shevach, E.M. Mechanisms of foxp3+ T regulatory cell-mediated suppression. Immunity 2009, 30, 636–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klebanoff, C.A.; Gattinoni, L.; Restifo, N.P. Sorting through subsets: Which T-cell populations mediate highly effective adoptive immunotherapy? J. Immunother. (Hagerstown MD 1997) 2012, 35, 651–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robbins, P.F.; Kassim, S.H.; Tran, T.L.N.; Crystal, J.S.; Morgan, R.A.; Feldman, S.A.; Yang, J.C.; Dudley, M.E.; Wunderlich, J.R.; Sherry, R.M.; et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: Long-term follow-up and correlates with response. Clin. Cancer Res. 2015, 21, 1019–1027. [Google Scholar] [CrossRef] [Green Version]
- Hoffmann, J.-M.; Schubert, M.-L.; Wang, L.; Hückelhoven, A.; Sellner, L.; Stock, S.; Schmitt, A.; Kleist, C.; Gern, U.; Loskog, A.; et al. Differences in Expansion Potential of Naive Chimeric Antigen Receptor T Cells from Healthy Donors and Untreated Chronic Lymphocytic Leukemia Patients. Front. Immunol. 2018, 8, 1956. [Google Scholar] [CrossRef] [Green Version]
- Cieri, N.; Camisa, B.; Cocchiarella, F.; Forcato, M.; Oliveira, G.; Provasi, E.; Bondanza, A.; Bordignon, C.; Peccatori, J.; Ciceri, F.; et al. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood 2013, 121, 573–584. [Google Scholar] [CrossRef]
- Xu, Y.; Zhang, M.; Ramos, C.A.; Durett, A.; Liu, E.; Dakhova, O.; Liu, H.; Creighton, C.J.; Gee, A.P.; Heslop, H.E.; et al. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood 2014, 123, 3750–3759. [Google Scholar] [CrossRef] [Green Version]
- Santegoets, S.J.A.M.; Turksma, A.W.; Suhoski, M.M.; Stam, A.G.M.; Albelda, S.M.; Hooijberg, E.; Scheper, R.J.; van den Eertwegh, A.J.M.; Gerritsen, W.R.; Powell, D.J.; et al. IL-21 promotes the expansion of CD27+ CD28+ tumor infiltrating lymphocytes with high cytotoxic potential and low collateral expansion of regulatory T cells. J. Transl. Med. 2013, 11, 37. [Google Scholar] [CrossRef] [Green Version]
- Gattinoni, L.; Klebanoff, C.A.; Restifo, N.P. Paths to stemness: Building the ultimate antitumour T cell. Nat. Rev. Cancer 2012, 12, 671–684. [Google Scholar] [CrossRef]
- Schmueck-Henneresse, M.; Omer, B.; Shum, T.; Tashiro, H.; Mamonkin, M.; Lapteva, N.; Sharma, S.; Rollins, L.; Dotti, G.; Reinke, P.; et al. Comprehensive Approach for Identifying the T Cell Subset Origin of CD3 and CD28 Antibody-Activated Chimeric Antigen Receptor-Modified T Cells. J. Immunol. 2017, 199, 348–362. [Google Scholar] [CrossRef] [Green Version]
- Klebanoff, C.A.; Finkelstein, S.E.; Surman, D.R.; Lichtman, M.K.; Gattinoni, L.; Theoret, M.R.; Grewal, N.; Spiess, P.J.; Antony, P.A.; Palmer, D.C.; et al. IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8 + T Cells. Proc. Natl. Acad. Sci. USA 2004, 101, 1969–1974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hinrichs, C.S.; Spolski, R.; Paulos, C.M.; Gattinoni, L.; Kerstann, K.W.; Palmer, D.C.; Klebanoff, C.A.; Rosenberg, S.A.; Leonard, W.J.; Restifo, N.P. IL-2 and IL-21 confer opposing differentiation programs to CD8+ T cells for adoptive immunotherapy. Blood 2008, 111, 5326–5333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gattinoni, L.; Klebanoff, C.A.; Restifo, N.P. Pharmacologic induction of CD8+ T cell memory: Better living through chemistry. Sci. Transl. Med. 2009, 1, 11ps12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gattinoni, L.; Zhong, X.-S.; Palmer, D.C.; Ji, Y.; Hinrichs, C.S.; Yu, Z.; Wrzesinski, C.; Boni, A.; Cassard, L.; Garvin, L.M.; et al. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat. Med. 2009, 15, 808–813. [Google Scholar] [CrossRef]
- Petersen, C.T.; Hassan, M.; Morris, A.B.; Jeffery, J.; Lee, K.; Jagirdar, N.; Staton, A.D.; Raikar, S.S.; Spencer, H.T.; Sulchek, T.; et al. Improving T-cell expansion and function for adoptive T-cell therapy using ex vivo treatment with PI3Kδ inhibitors and VIP antagonists. Blood Adv. 2018, 2, 210–223. [Google Scholar] [CrossRef] [Green Version]
- Araki, K.; Turner, A.P.; Shaffer, V.O.; Gangappa, S.; Keller, S.A.; Bachmann, M.F.; Larsen, C.P.; Ahmed, R. mTOR regulates memory CD8 T-cell differentiation. Nature 2009, 460, 108–112. [Google Scholar] [CrossRef] [Green Version]
- Shan, Q.; Hu, S.; Chen, X.; Danahy, D.B.; Badovinac, V.P.; Zang, C.; Xue, H.-H. Ectopic Tcf1 expression instills a stem-like program in exhausted CD8+ T cells to enhance viral and tumor immunity. Cell Mol. Immunol. 2020, 1–16. [Google Scholar] [CrossRef]
- Page, N.; Klimek, B.; Roo, M.D.; Steinbach, K.; Soldati, H.; Lemeille, S.; Wagner, I.; Kreutzfeldt, M.; Liberto, G.D.; Vincenti, I.; et al. Expression of the DNA-Binding Factor TOX Promotes the Encephalitogenic Potential of Microbe-Induced Autoreactive CD8+ T Cells. Immunity 2019, 50, 763. [Google Scholar] [CrossRef]
- Wilkie, S.; van Schalkwyk, M.C.I.; Hobbs, S.; Davies, D.M.; van der Stegen, S.J.C.; Pereira, A.C.P.; Burbridge, S.E.; Box, C.; Eccles, S.A.; Maher, J. Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling. J. Clin. Immunol. 2012, 32, 1059–1070. [Google Scholar] [CrossRef]
- Kloss, C.C.; 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, 71–75. [Google Scholar] [CrossRef]
- Lanitis, E.; Poussin, M.; Klattenhoff, A.W.; Song, D.; Sandaltzopoulos, R.; June, C.H.; Powell, D.J. Chimeric antigen receptor T Cells with dissociated signaling domains exhibit focused antitumor activity with reduced potential for toxicity in vivo. Cancer Immunol. Res. 2013, 1, 43–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grada, Z.; Hegde, M.; Byrd, T.; Shaffer, D.R.; Ghazi, A.; Brawley, V.S.; Corder, A.; Schönfeld, K.; Koch, J.; Dotti, G.; et al. TanCAR: A Novel Bispecific Chimeric Antigen Receptor for Cancer Immunotherapy. Mol. Ther. Nucleic Acids 2013, 2, e105. [Google Scholar] [CrossRef] [PubMed]
- Hegde, M.; Mukherjee, M.; Grada, Z.; Pignata, A.; Landi, D.; Navai, S.A.; Wakefield, A.; Fousek, K.; Bielamowicz, K.; Chow, K.K.H.; et al. Tandem CAR T cells targeting HER2 and IL13Rα2 mitigate tumor antigen escape. J. Clin. Investig. 2016, 126, 3036–3052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zah, E.; Lin, M.-Y.; Silva-Benedict, A.; Jensen, M.C.; Chen, Y.Y. T Cells Expressing CD19/CD20 Bispecific Chimeric Antigen Receptors Prevent Antigen Escape by Malignant B Cells. Cancer Immunol. Res. 2016, 4, 498–508. [Google Scholar] [CrossRef] [Green Version]
- Munter, S.D.; Ingels, J.; Goetgeluk, G.; Bonte, S.; Pille, M.; Weening, K.; Kerre, T.; Abken, H.; Vandekerckhove, B. Nanobody Based Dual Specific CARs. Int. J. Mol. Sci. 2018, 19, 403. [Google Scholar] [CrossRef] [Green Version]
- Fedorov, V.D.; Themeli, M.; Sadelain, M. PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses. Sci. Transl. Med. 2013, 5, 215ra172. [Google Scholar] [CrossRef] [Green Version]
- Morsut, L.; Roybal, K.T.; Xiong, X.; Gordley, R.M.; Coyle, S.M.; Thomson, M.; Lim, W.A. Engineering Customized Cell Sensing and Response Behaviors Using Synthetic Notch Receptors. Cell 2016, 164, 780–791. [Google Scholar] [CrossRef] [Green Version]
- Roybal, K.T.; Williams, J.Z.; Morsut, L.; Rupp, L.J.; Kolinko, I.; Choe, J.H.; Walker, W.J.; McNally, K.A.; Lim, W.A. Engineering T Cells with Customized Therapeutic Response Programs Using Synthetic Notch Receptors. Cell 2016, 167, 419–432.e16. [Google Scholar] [CrossRef] [Green Version]
- Roybal, K.T.; Rupp, L.J.; Morsut, L.; Walker, W.J.; McNally, K.A.; Park, J.S.; Lim, W.A. Precision Tumor Recognition by T Cells With Combinatorial Antigen-Sensing Circuits. Cell 2016, 164, 770–779. [Google Scholar] [CrossRef] [Green Version]
- Srivastava, S.; Salter, A.I.; Liggitt, D.; Yechan-Gunja, S.; Sarvothama, M.; Cooper, K.; Smythe, K.S.; Dudakov, J.A.; Pierce, R.H.; Rader, C.; et al. Logic-Gated ROR1 Chimeric Antigen Receptor Expression Rescues T Cell-Mediated Toxicity to Normal Tissues and Enables Selective Tumor Targeting. Cancer Cell 2019, 35, 489–503.e8. [Google Scholar] [CrossRef] [Green Version]
- Nabet, B.; Roberts, J.M.; Buckley, D.L.; Paulk, J.; Dastjerdi, S.; Yang, A.; Leggett, A.L.; Erb, M.A.; Lawlor, M.A.; Souza, A.; et al. The dTAG system for immediate and target-specific protein degradation. Nat. Chem. Biol. 2018, 14, 431–441. [Google Scholar] [CrossRef] [PubMed]
- Fisher, J.; Abramowski, P.; Don, N.D.W.; Flutter, B.; Capsomidis, A.; Cheung, G.W.-K.; Gustafsson, K.; Anderson, J. Avoidance of On-Target Off-Tumor Activation Using a Co-stimulation-Only Chimeric Antigen Receptor. Mol. Ther. 2017, 25, 1234–1247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rosenberg, S.A. IL-2: The First Effective Immunotherapy for Human Cancer. J. Immunol. 2014, 192, 5451–5458. [Google Scholar] [CrossRef] [PubMed]
- Yee, C.; Thompson, J.A.; Byrd, D.; Riddell, S.R.; Roche, P.; Celis, E.; Greenberg, P.D. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: In vivo persistence, migration, and antitumor effect of transferred T cells. Proc. Natl. Acad. Sci. USA 2002, 99, 16168–16173. [Google Scholar] [CrossRef] [Green Version]
- Mitsuyasu, R.T.; Anton, P.A.; Deeks, S.G.; Scadden, D.T.; Connick, E.; Downs, M.T.; Bakker, A.; Roberts, M.R.; June, C.H.; Jalali, S.; et al. Prolonged survival and tissue trafficking following adoptive transfer of CD4zeta gene-modified autologous CD4(+) and CD8(+) T cells in human immunodeficiency virus-infected subjects. Blood 2000, 96, 785–793. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Chua, K.S.; Guimond, M.; Kapoor, V.; Brown, M.V.; Fleisher, T.A.; Long, L.M.; Bernstein, D.; Hill, B.J.; Douek, D.C.; et al. Lymphopenia and interleukin-2 therapy alter homeostasis of CD4+CD25+ regulatory T cells. Nat. Med. 2005, 11, 1238–1243. [Google Scholar] [CrossRef]
- Cohen, J. IL-12 Deaths: Explanation and a Puzzle. Science 1995, 270, 908. [Google Scholar] [CrossRef]
- Leen, A.M.; Sukumaran, S.; Watanabe, N.; Mohammed, S.; Keirnan, J.; Yanagisawa, R.; Anurathapan, U.; Rendon, D.; Heslop, H.E.; Rooney, C.M.; et al. Reversal of tumor immune inhibition using a chimeric cytokine receptor. Mol. Ther. J. Am. Soc. Gene Ther. 2014, 22, 1211–1220. [Google Scholar] [CrossRef] [Green Version]
- Mohammed, S.; Sukumaran, S.; Bajgain, P.; Watanabe, N.; Heslop, H.E.; Rooney, C.M.; Brenner, M.K.; Fisher, W.E.; Leen, A.M.; Vera, J.F. Improving Chimeric Antigen Receptor-Modified T Cell Function by Reversing the Immunosuppressive Tumor Microenvironment of Pancreatic Cancer. Mol. Ther. 2017, 25, 249–258. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Jiang, H.; Luo, H.; Sun, Y.; Shi, B.; Sun, R.; Li, Z. An IL-4/21 Inverted Cytokine Receptor Improving CAR-T Cell Potency in Immunosuppressive Solid-Tumor Microenvironment. Front. Immunol. 2019, 10, 1691. [Google Scholar] [CrossRef] [Green Version]
- Pegram, H.J.; Lee, J.C.; Hayman, E.G.; Imperato, G.H.; Tedder, T.F.; Sadelain, M.; Brentjens, R.J. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood 2012, 119, 4133–4141. [Google Scholar] [CrossRef] [PubMed]
- Chmielewski, M.; Kopecky, C.; Hombach, A.A.; Abken, H. IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively Muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer Res. 2011, 71, 5697–5706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koneru, M.; O’Cearbhaill, R.; Pendharkar, S.; Spriggs, D.R.; Brentjens, R.J. A phase I clinical trial of adoptive T cell therapy using IL-12 secreting MUC-16(ecto) directed chimeric antigen receptors for recurrent ovarian cancer. J. Transl. Med. 2015, 13, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, B.; Ren, J.; Luo, Y.; Keith, B.; Young, R.M.; Scholler, J.; Zhao, Y.; June, C.H. Augmentation of Antitumor Immunity by Human and Mouse CAR T Cells Secreting IL-18. Cell Rep. 2017, 20, 3025–3033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hurton, L.V.; Singh, H.; Najjar, A.M.; Switzer, K.C.; Mi, T.; Maiti, S.; Olivares, S.; Rabinovich, B.; Huls, H.; Forget, M.-A.; et al. Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells. Proc. Natl. Acad. Sci. USA 2016, 113, E7788–E7797. [Google Scholar] [CrossRef] [Green Version]
- Boice, M.; Salloum, D.; Mourcin, F.; Sanghvi, V.; Amin, R.; Oricchio, E.; Jiang, M.; Mottok, A.; Denis-Lagache, N.; Ciriello, G.; et al. Loss of the HVEM Tumor Suppressor in Lymphoma and Restoration by Modified CAR-T Cells. Cell 2016, 167, 405–418.e13. [Google Scholar] [CrossRef] [Green Version]
- Kagoya, Y.; Tanaka, S.; Guo, T.; Anczurowski, M.; Wang, C.-H.; Saso, K.; Butler, M.O.; Minden, M.D.; Hirano, N. A novel chimeric antigen receptor containing a JAK-STAT signaling domain mediates superior antitumor effects. Nat. Med. 2018, 24, 352–359. [Google Scholar] [CrossRef]
- Spranger, S.; Gajewski, T.F. Impact of oncogenic pathways on evasion of antitumour immune responses. Nat. Rev. Cancer 2018, 18, 139–147. [Google Scholar] [CrossRef]
- Kloss, C.C.; Lee, J.; Zhang, A.; Chen, F.; Melenhorst, J.J.; Lacey, S.F.; Maus, M.V.; Fraietta, J.A.; Zhao, Y.; June, C.H. Dominant-Negative TGF-β Receptor Enhances PSMA-Targeted Human CAR T Cell Proliferation and Augments Prostate Cancer Eradication. Mol. Ther. 2018, 26, 1855–1866. [Google Scholar] [CrossRef] [Green Version]
- John, L.B.; Devaud, C.; Duong, C.P.M.; Yong, C.S.; Beavis, P.A.; Haynes, N.M.; Chow, M.T.; Smyth, M.J.; Kershaw, M.H.; Darcy, P.K. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin. Cancer Res. 2013, 19, 5636–5646. [Google Scholar] [CrossRef] [Green Version]
- Cherkassky, L.; Morello, A.; Villena-Vargas, J.; Feng, Y.; Dimitrov, D.S.; Jones, D.R.; Sadelain, M.; Adusumilli, P.S. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J. Clin. Investig. 2016, 126, 3130–3144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rupp, L.J.; Schumann, K.; Roybal, K.T.; Gate, R.E.; Ye, C.J.; Lim, W.A.; Marson, A. CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells. Sci. Rep. UK 2017, 7, 737. [Google Scholar] [CrossRef] [PubMed]
- Chong, E.A.; Melenhorst, J.J.; Lacey, S.F.; Ambrose, D.E.; Gonzalez, V.; Levine, B.; June, C.H.; Schuster, S.J. PD-1 Blockade Modulates Chimeric Antigen Receptor (CAR) Modified T Cells and Induces Tumor Regression: Refueling the CAR. Blood 2016, 129, 1039–1041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ren, J.; Liu, X.; Fang, C.; Jiang, S.; June, C.H.; Zhao, Y. Multiplex Genome Editing to Generate Universal CAR T Cells Resistant to PD1 Inhibition. Clin. Cancer Res. 2016, 23, 2255–2266. [Google Scholar] [CrossRef] [Green Version]
- Heczey, A.; Louis, C.U.; Savoldo, B.; Dakhova, O.; Durett, A.; Grilley, B.; Liu, H.; Wu, M.F.; Mei, Z.; Gee, A.; et al. CAR T Cells Administered in Combination with Lymphodepletion and PD-1 Inhibition to Patients with Neuroblastoma. Mol. Ther. 2017, 25. [Google Scholar] [CrossRef] [Green Version]
- Stadtmauer, E.A.; Fraietta, J.A.; Davis, M.M.; Cohen, A.D.; Weber, K.L.; Lancaster, E.; Mangan, P.A.; Kulikovskaya, I.; Gupta, M.; Chen, F.; et al. CRISPR-engineered T cells in patients with refractory cancer. Science N. Y. 2020, 367, eaba7365. [Google Scholar] [CrossRef] [PubMed]
- Qasim, W.; Zhan, H.; Samarasinghe, S.; Adams, S.; Amrolia, P.; Stafford, S.; Butler, K.; Rivat, C.; Wright, G.; Somana, K.; et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci. Transl. Med. 2017, 9, eaaj2013. [Google Scholar] [CrossRef]
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Gavriil, A.; Barisa, M.; Halliwell, E.; Anderson, J. Engineering Solutions for Mitigation of Chimeric Antigen Receptor T-Cell Dysfunction. Cancers 2020, 12, 2326. https://doi.org/10.3390/cancers12082326
Gavriil A, Barisa M, Halliwell E, Anderson J. Engineering Solutions for Mitigation of Chimeric Antigen Receptor T-Cell Dysfunction. Cancers. 2020; 12(8):2326. https://doi.org/10.3390/cancers12082326
Chicago/Turabian StyleGavriil, Artemis, Marta Barisa, Emma Halliwell, and John Anderson. 2020. "Engineering Solutions for Mitigation of Chimeric Antigen Receptor T-Cell Dysfunction" Cancers 12, no. 8: 2326. https://doi.org/10.3390/cancers12082326
APA StyleGavriil, A., Barisa, M., Halliwell, E., & Anderson, J. (2020). Engineering Solutions for Mitigation of Chimeric Antigen Receptor T-Cell Dysfunction. Cancers, 12(8), 2326. https://doi.org/10.3390/cancers12082326