1932

Abstract

The field of cancer immunotherapy has been re-energized by the application of chimeric antigen receptor (CAR) T cell therapy in cancers. These CAR T cells are engineered to express synthetic receptors that redirect polyclonal T cells to surface antigens for subsequent tumor elimination. Many CARs are designed with elements that augment T cell persistence and activity. To date, CAR T cells have demonstrated tremendous success in eradicating hematologic malignancies (e.g., CD19 CARs in leukemias). However, this success has yet to be extrapolated to solid tumors, and the reasons for this are being actively investigated. We characterize some of the challenges that CAR T cells have to surmount in the solid tumor microenvironment and new approaches that are being considered to overcome these hurdles.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-062315-120245
2017-01-14
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/med/68/1/annurev-med-062315-120245.html?itemId=/content/journals/10.1146/annurev-med-062315-120245&mimeType=html&fmt=ahah

Literature Cited

  1. Topalian SL, Drake CG, Pardoll DM. 1.  2015. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 27:450–61 [Google Scholar]
  2. Willemsen RA, Debets R, Hart E. 2.  et al. 2001. A phage display selected fab fragment with MHC class I-restricted specificity for MAGE-A1 allows for retargeting of primary human T lymphocytes. Gene Ther 8:1601–8 [Google Scholar]
  3. Gill S, Maus MV, Porter DL. 3.  2015. Chimeric antigen receptor T cell therapy: 25 years in the making. Blood Rev 30:157–67 [Google Scholar]
  4. Fousek K, Ahmed N. 4.  2015. The evolution of T-cell therapies for solid malignancies. Clin. Cancer Res. 21:3384–92 [Google Scholar]
  5. Louis CU, Savoldo B, Dotti G. 5.  et al. 2011. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 118:6050–56 [Google Scholar]
  6. Ahmed N, Brawley VS, Hegde M. 6.  et al. 2015. Human epidermal growth factor receptor 2 (HER2)–specific chimeric antigen receptor-modified T cells for the immunotherapy of HER2-positive sarcoma. J. Clin. Oncol. 33:1688–96 [Google Scholar]
  7. Feng K, Guo Y, Dai H. 7.  et al. 2016. Chimeric antigen receptor-modified T cells for the immunotherapy of patients with EGFR-expressing advanced relapsed/refractory non-small cell lung cancer. Sci. China Life Sci. 59:468–7 [Google Scholar]
  8. Restifo NP, Dudley ME, Rosenberg SA. 8.  2012. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat. Rev. Immunol. 12:269–81 [Google Scholar]
  9. Kenter GG, Welters MJ, Valentijn AR. 9.  et al. 2009. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N. Engl. J. Med. 361:1838–47 [Google Scholar]
  10. Johnson LA, Scholler J, Ohkuri T. 10.  et al. 2015. Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci. Transl. Med. 7:275ra22 [Google Scholar]
  11. Morgan RA, Johnson LA, Davis JL. 11.  et al. 2012. Recognition of glioma stem cells by genetically modified T cells targeting EGFRvIII and development of adoptive cell therapy for glioma. Hum. Gene Ther. 23:1043–53 [Google Scholar]
  12. Wilkie S, Picco G, Foster J. 12.  et al. 2008. Retargeting of human T cells to tumor-associated MUC1: the evolution of a chimeric antigen receptor. J. Immunol. 180:4901–9 [Google Scholar]
  13. Chekmasova AA, Rao TD, Nikhamin Y. 13.  et al. 2010. Successful eradication of established peritoneal ovarian tumors in SCID-Beige mice following adoptive transfer of T cells genetically targeted to the MUC16 antigen. Clin. Cancer Res. 16:3594–606 [Google Scholar]
  14. Morello A, Sadelain M, Adusumilli PS. 14.  2015. Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov 6:133–46 [Google Scholar]
  15. Pastan I, Hassan R. 15.  2014. Discovery of mesothelin and exploiting it as a target for immunotherapy. Cancer Res 74:2907–12 [Google Scholar]
  16. Chowdhury PS, Viner JL, Beers R, Pastan I. 16.  1998. Isolation of a high-affinity stable single-chain Fv specific for mesothelin from DNA-immunized mice by phage display and construction of a recombinant immunotoxin with anti-tumor activity. PNAS 95:669–74 [Google Scholar]
  17. Lanitis E, Poussin M, Hagemann IS. 17.  et al. 2012. Redirected antitumor activity of primary human lymphocytes transduced with a fully human anti-mesothelin chimeric receptor. Mol. Ther. 20:633–43 [Google Scholar]
  18. Adusumilli PS, Cherkassky L, Villena-Vargas J. 18.  et al. 2014. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci. Transl. Med. 6:261ra151 [Google Scholar]
  19. Morgan RA, Yang JC, Kitano M. 19.  et al. 2010. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 18:843–51 [Google Scholar]
  20. Lanitis E, Poussin M, Klattenhoff AW. 20.  et al. 2013. Chimeric antigen receptor T cells with dissociated signaling domains exhibit focused antitumor activity with reduced potential for toxicity in vivo. Cancer Immunol. Res. 1:43–53 [Google Scholar]
  21. Kloss CC, Condomines M, Cartellieri M. 21.  et al. 2013. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat. Biotechnol. 31:71–75 [Google Scholar]
  22. Sampson JH, Choi BD, Sanchez-Perez L. 22.  et al. 2014. EGFRvIII mCAR-modified T-cell therapy cures mice with established intracerebral glioma and generates host immunity against tumor-antigen loss. Clin. Cancer Res. 20:972–84 [Google Scholar]
  23. Beatty GL, Haas AR, Maus MV. 23.  et al. 2014. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol. Res. 2:112–20 [Google Scholar]
  24. Wilkie S, van Schalkwyk MC, Hobbs S. 24.  et al. 2012. Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling. J. Clin. Immunol. 32:1059–70 [Google Scholar]
  25. Grada Z, Hegde M, Byrd T. 25.  et al. 2013. TanCAR: a novel bispecific chimeric antigen receptor for cancer immunotherapy. Mol. Ther. Nucleic Acids 2:e105 [Google Scholar]
  26. Wang LC, Lo A, Scholler J. 26.  et al. 2014. Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicity. Cancer Immunol. Res. 2:154–66 [Google Scholar]
  27. Chinnasamy D, Tran E, Yu Z. 27.  et al. 2013. Simultaneous targeting of tumor antigens and the tumor vasculature using T lymphocyte transfer synergize to induce regression of established tumors in mice. Cancer Res 73:3371–80 [Google Scholar]
  28. Chmielewski M, Hombach AA, Abken H. 28.  2014. Of CARs and TRUCKs: chimeric antigen receptor (CAR) T cells engineered with an inducible cytokine to modulate the tumor stroma. Immunol. Rev. 257:83–90 [Google Scholar]
  29. Zhang L, Morgan RA, Beane JD. 29.  et al. 2015. Tumor-infiltrating lymphocytes genetically engineered with an inducible gene encoding interleukin-12 for the immunotherapy of metastatic melanoma. Clin. Cancer Res. 21:2278–88 [Google Scholar]
  30. Pegram HJ, Lee JC, Hayman EG. 30.  et al. 2012. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood 119:4133–41 [Google Scholar]
  31. Iwahori K, Kakarla S, Velasquez MP. 31.  et al. 2015. Engager T cells: a new class of antigen-specific T cells that redirect bystander T cells. Mol. Ther. 23:171–78 [Google Scholar]
  32. Compte M, Blanco B, Serrano F. 32.  et al. 2007. Inhibition of tumor growth in vivo by in situ secretion of bispecific anti-CEA × anti-CD3 diabodies from lentivirally transduced human lymphocytes. Cancer Gene Ther 14:380–88 [Google Scholar]
  33. Curran KJ, Seinstra BA, Nikhamin Y. 33.  et al. 2015. Enhancing antitumor efficacy of chimeric antigen receptor T cells through constitutive CD40L expression. Mol. Ther. 23:769–78 [Google Scholar]
  34. Harlin H, Meng Y, Peterson AC. 34.  et al. 2009. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res 69:3077–85 [Google Scholar]
  35. Craddock JA, Lu A, Bear A. 35.  et al. 2010. Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b. J. Immunother. 33:780–88 [Google Scholar]
  36. Moon EK, Carpenito C, Sun J. 36.  et al. 2011. Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptor. Clin. Cancer Res. 17:4719–30 [Google Scholar]
  37. Newick K, O'Brien S, Sun J. 37.  et al. 2016. Augmentation of CAR T cell trafficking and antitumor efficacy by blocking protein kinase A (PKA) localization. Cancer Immunol. Res. 4:541–51 [Google Scholar]
  38. Nishio N, Diaconu I, Liu H. 38.  et al. 2014. Armed oncolytic virus enhances immune functions of chimeric antigen receptor-modified T cells in solid tumors. Cancer Res 74:5195–205 [Google Scholar]
  39. VanSeggelen H, Hammill JA, Dvorkin-Gheva A. 39.  et al. 2015. T cells engineered with chimeric antigen receptors targeting NKG2D ligands display lethal toxicity in mice. Mol. Ther. 23:1600–10 [Google Scholar]
  40. Caruana I, Savoldo B, Hoyos V. 40.  et al. 2015. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat. Med. 21:524–29 [Google Scholar]
  41. Zhang Y, Ertl HC. 41.  2016. Starved and asphyxiated: How can CD8+ T cells within a tumor microenvironment prevent tumor progression?. Front. Immunol. 7:32 [Google Scholar]
  42. Hatfield SM, Kjaergaard J, Lukashev D. 42.  et al. 2015. Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci. Transl. Med. 7:277ra30 [Google Scholar]
  43. Fischer K, Hoffmann P, Voelkl S. 43.  et al. 2007. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 109:3812–19 [Google Scholar]
  44. Jacobs SR, Herman CE, Maciver NJ. 44.  et al. 2008. Glucose uptake is limiting in T cell activation and requires CD28-mediated Akt-dependent and independent pathways. J. Immunol. 180:4476–86 [Google Scholar]
  45. Howie D, Waldmann H, Cobbold S. 45.  2014. Nutrient sensing via mTOR in T cells maintains a tolerogenic microenvironment. Front. Immunol. 5:409 [Google Scholar]
  46. Ninomiya S, Narala N, Huye L. 46.  et al. 2015. Tumor indoleamine 2,3-dioxygenase (IDO) inhibits CD19-CAR T cells and is downregulated by lymphodepleting drugs. Blood 125:3905–16 [Google Scholar]
  47. Velica P, Zech M, Henson S. 47.  et al. 2015. Genetic regulation of fate decisions in therapeutic T cells to enhance tumor protection and memory formation. Cancer Res 75:2641–52 [Google Scholar]
  48. Sukumar M, Liu J, Ji Y. 48.  et al. 2013. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J. Clin. Investig. 123:4479–88 [Google Scholar]
  49. Goodwin JS, Bankhurst AD, Messner RP. 49.  1977. Suppression of human T-cell mitogenesis by prostaglandin. Existence of a prostaglandin-producing suppressor cell. J. Exp. Med. 146:1719–34 [Google Scholar]
  50. Su Y, Huang X, Raskovalova T. 50.  et al. 2008. Cooperation of adenosine and prostaglandin E2 (PGE2) in amplification of cAMP-PKA signaling and immunosuppression. Cancer Immunol. Immunother. 57:1611–23 [Google Scholar]
  51. Oh SA, Li MO. 51.  2013. TGF-beta: guardian of T cell function. J. Immunol. 191:3973–79 [Google Scholar]
  52. Massague J. 52.  2008. TGFβ in cancer. Cell 134:215–30 [Google Scholar]
  53. Wallace A, Kapoor V, Sun J. 53.  et al. 2008. Transforming growth factor-beta receptor blockade augments the effectiveness of adoptive T-cell therapy of established solid cancers. Clin. Cancer Res. 14:3966–74 [Google Scholar]
  54. Bollard CM, Rossig C, Calonge MJ. 54.  et al. 2002. Adapting a transforming growth factor beta-related tumor protection strategy to enhance antitumor immunity. Blood 99:3179–87 [Google Scholar]
  55. Gabrilovich DI, Nagaraj S. 55.  2009. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 9:162–74 [Google Scholar]
  56. Mussai F, Egan S, Hunter S. 56.  et al. 2015. Neuroblastoma arginase activity creates an immunosuppressive microenvironment that impairs autologous and engineered immunity. Cancer Res 75:3043–53 [Google Scholar]
  57. Burga RA, Thorn M, Point GR. 57.  et al. 2015. Liver myeloid-derived suppressor cells expand in response to liver metastases in mice and inhibit the anti-tumor efficacy of anti-CEA CAR-T. Cancer Immunol. Immunother. 64:817–29 [Google Scholar]
  58. Spear P, Barber A, Rynda-Apple A, Sentman CL. 58.  2012. Chimeric antigen receptor T cells shape myeloid cell function within the tumor microenvironment through IFN-gamma and GM-CSF. J. Immunol. 188:6389–98 [Google Scholar]
  59. Sakaguchi S, Miyara M, Costantino CM, Hafler DA. 59.  2010. FOXP3+ regulatory T cells in the human immune system. Nat. Rev. Immunol. 10:490–500 [Google Scholar]
  60. Zhou Q, Munger ME, Highfill SL. 60.  et al. 2010. Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia. Blood 116:2484–93 [Google Scholar]
  61. Liu Y, Wang L, Predina J. 61.  et al. 2013. Inhibition of p300 impairs Foxp3+ T regulatory cell function and promotes antitumor immunity. Nat. Med. 19:1173–77 [Google Scholar]
  62. Perna SK, Pagliara D, Mahendravada A. 62.  et al. 2014. Interleukin-7 mediates selective expansion of tumor-redirected cytotoxic T lymphocytes (CTLs) without enhancement of regulatory T-cell inhibition. Clin. Cancer Res. 20:131–39 [Google Scholar]
  63. Yao X, Ahmadzadeh M, Lu YC. 63.  et al. 2012. Levels of peripheral CD4+FoxP3+ regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancer. Blood 119:5688–96 [Google Scholar]
  64. Kofler DM, Chmielewski M, Rappl G. 64.  et al. 2011. CD28 costimulation impairs the efficacy of a redirected T-cell antitumor attack in the presence of regulatory T cells which can be overcome by preventing Lck activation. Mol. Ther. 19:760–67 [Google Scholar]
  65. Markley JC, Sadelain M. 65.  2010. IL-7 and IL-21 are superior to IL-2 and IL-15 in promoting human T cell-mediated rejection of systemic lymphoma in immunodeficient mice. Blood 115:3508–19 [Google Scholar]
  66. Finney HM, Akbar AN, Lawson AD. 66.  2004. Activation of resting human primary T cells with chimeric receptors: costimulation from CD28, inducible costimulator, CD134, and CD137 in series with signals from the TCR zeta chain. J. Immunol. 172:104–13 [Google Scholar]
  67. Moon EK, Wang LC, Dolfi DV. 67.  et al. 2014. Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor-transduced human T cells in solid tumors. Clin. Cancer Res. 20:4262–73 [Google Scholar]
  68. John LB, Devaud C, Duong CP. 68.  et al. 2013. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin. Cancer Res. 19:5636–46 [Google Scholar]
  69. Moon EK, Ranganathan R, Eruslanov E. 69.  et al. 2016. Blockade of programmed death 1 augments the ability of human T cells engineered to target NY-ESO-1 to control tumor growth after adoptive transfer. Clin. Cancer Res. 22:436–47 [Google Scholar]
  70. Kobold S, Grassmann S, Chaloupka M. 70.  et al. 2015. Impact of a new fusion receptor on PD-1-mediated immunosuppression in adoptive T cell therapy. J. Natl. Cancer Inst. 107:djv146 [Google Scholar]
  71. Mahvi DA, Meyers JV, Tatar AJ. 71.  et al. 2015. CTLA-4 blockade plus adoptive T-cell transfer promotes optimal melanoma immunity in mice. J. Immunother. 38:54–61 [Google Scholar]
  72. Wang LC, Riese MJ, Moon EK, Albelda SM. 72.  2013. Overcoming intrinsic inhibitory pathways to augment the antineoplastic activity of adoptively transferred T cells: re-tuning your CAR before hitting a rocky road. Oncoimmunology 2:e26492 [Google Scholar]
  73. Riese MJ, Wang LC, Moon EK. 73.  et al. 2013. Enhanced effector responses in activated CD8+ T cells deficient in diacylglycerol kinases. Cancer Res 73:3566–77 [Google Scholar]
  74. Charo J, Finkelstein SE, Grewal N. 74.  et al. 2005. Bcl-2 overexpression enhances tumor-specific T-cell survival. Cancer Res 65:2001–8 [Google Scholar]
  75. Guedan S, Chen X, Madar A. 75.  et al. 2014. ICOS-based chimeric antigen receptors program bipolar TH17/TH1 cells. Blood 124:1070–80 [Google Scholar]
  76. Song DG, Ye Q, Poussin M. 76.  et al. 2012. CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivo. Blood 119:696–706 [Google Scholar]
  77. Wang E, Wang LC, Tsai CY. 77.  et al. 2015. Generation of potent T-cell immunotherapy for cancer using DAP12-based, multichain, chimeric immunoreceptors. Cancer Immunol. Res. 3:815–26 [Google Scholar]
/content/journals/10.1146/annurev-med-062315-120245
Loading
/content/journals/10.1146/annurev-med-062315-120245
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error