Download REFERENCES

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
REFERENCES
1. Rosenberg, S. A. & Restifo, N. P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348, 62–68 (2015).
2. Dudley, M. E., Wunderlich, J. R., Shelton, T. E., Even, J. & Rosenberg, S. A. Generation of tumor-infiltrating lymphocyte cultures for use in
adoptive transfer therapy for melanoma patients. J. Immunother. Hagerstown Md 1997 26, 332 (2003).
3. Rosenberg, S. A. et al. Durable Complete Responses in Heavily Pretreated Patients with Metastatic Melanoma Using T-Cell Transfer
Immunotherapy. Clin. Cancer Res. 17, 4550–4557 (2011).
4. Kawalekar, O. U. et al. Distinct Signaling of Coreceptors Regulates Specific Metabolism Pathways and Impacts Memory Development in
CAR T Cells. Immunity 44, 380–390 (2016).
5. Zhao, Z. et al. Structural Design of Engineered Costimulation Determines Tumor Rejection Kinetics and Persistence of CAR T Cells.
Cancer Cell 28, 415–428 (2015).
6. Brentjens, R. J. et al. CD19-Targeted T Cells Rapidly Induce Molecular Remissions in Adults with Chemotherapy-Refractory Acute
Lymphoblastic Leukemia. Sci. Transl. Med. 5, 177ra38–177ra38 (2013).
7. Maude, S. L. et al. Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia. N. Engl. J. Med. 371, 1507–1517 (2014).
8. Lee, D. W. et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a
phase 1 dose-escalation trial. The Lancet 385, 517–528 (2015).
9. Maude, S. L., Teachey, D. T., Porter, D. L. & Grupp, S. A. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic
leukemia. Blood 125, 4017–4023 (2015).
10. Pule, M. A. et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in
individuals with neuroblastoma. Nat. Med. 14, 1264–1270 (2008).
11. Figueroa, J. A. et al. Chimeric antigen receptor engineering: a right step in the evolution of adoptive cellular immunotherapy. Int. Rev.
Immunol. 34, 154–187 (2015).
12. Brentjens, R. J. et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or
chemotherapy refractory B-cell leukemias. Blood 118, 4817–4828 (2011).
13. Morgan, R. A. et al. Case Report of a Serious Adverse Event Following the Administration of T Cells Transduced With a Chimeric Antigen
Receptor Recognizing ERBB2. Mol. Ther. 18, 843–851 (2010).
14. Ahmed, N. et al. 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, 1688–1696 (2015).
15. Dotti, G., Gottschalk, S., Savoldo, B. & Brenner, M. K. Design and development of therapies using chimeric antigen receptor-expressing T
cells. Immunol. Rev. 257, 107–126 (2014).
16. Gargett, T. & Brown, M. P. Different cytokine and stimulation conditions influence the expansion and immune phenotype of thirdgeneration chimeric antigen receptor T cells specific for tumor antigen GD2. Cytotherapy 17, 487–495 (2015).
17. Yang, S. et al. In vitro generated anti-tumor T lymphocytes exhibit distinct subsets mimicking in vivo antigen-experienced cells. Cancer
Immunol. Immunother. 60, 739–749 (2011).
18. Neeson, P. et al. Ex vivo culture of chimeric antigen receptor T cells generates functional CD8+ T cells with effector and central memorylike phenotype. Gene Ther. 17, 1105–1116 (2010).
19. Gattinoni, L. & Restifo, N. P. Moving T memory stem cells to the clinic. BLOOD 121, (2013).
20. June, C. H., Ledbetter, J. A., Linsley, P. S. & Thompson, C. B. Role of the CD28 receptor in T-cell activation. Immunol. Today 11, 211–216
(1990).
21. Porter, D. L. et al. A phase 1 trial of donor lymphocyte infusions expanded and activated ex vivo via CD3/CD28 costimulation. Blood 107,
1325–1331 (2006).
22. Gomez-Eerland, R. et al. Manufacture of Gene-Modified Human T-Cells with a Memory Stem/Central Memory Phenotype. Hum. Gene
Ther. Methods 25, 277–287 (2014).
23. Cieri, N. et al. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood 121, 573–584 (2013).
24. Xu, Y. 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 IL15. Blood 123, 3750–3759 (2014).
25. Jensen, M. C. et al. Antitransgene Rejection Responses Contribute to Attenuated Persistence of Adoptively Transferred CD20/CD19Specific Chimeric Antigen Receptor Redirected T Cells in Humans. Biol. Blood Marrow Transplant. 16, 1245–1256 (2010).
26. Savoldo, B. et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor–modified T cells in lymphoma
patients. J. Clin. Invest. 121, 1822–1826 (2011).
27. Kochenderfer, J. N. et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic
hematopoietic stem cell transplantation. Blood 122, 4129–4139 (2013).
28. Davila, M. L. et al. Efficacy and Toxicity Management of 19-28z CAR T Cell Therapy in B Cell Acute Lymphoblastic Leukemia. Sci. Transl.
Med. 6, 224ra25–224ra25 (2014).
29. Gross, G. & Eshhar, Z. Therapeutic Potential of T Cell Chimeric Antigen Receptors (CARs) in Cancer Treatment: Counteracting OffTumor Toxicities for Safe CAR T Cell Therapy. Annu. Rev. Pharmacol. Toxicol. 56, 59–83 (2016).
30. Dai, H., Wang, Y., Lu, X. & Han, W. Chimeric Antigen Receptors Modified T-Cells for Cancer Therapy. J. Natl. Cancer Inst. 108, djv439
(2016).
31. Lamers, C. H. et al. Treatment of Metastatic Renal Cell Carcinoma With CAIX CAR-engineered T cells: Clinical Evaluation and
Management of On-target Toxicity. Mol. Ther. 21, 904–912 (2013).
32. Park, J. R. et al. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with
neuroblastoma. Mol. Ther. J. Am. Soc. Gene Ther. 15, 825–833 (2007).
33. Louis, C. U. et al. Antitumor activity and long-term fate of chimeric antigen receptor–positive T cells in patients with neuroblastoma.
Blood 118, 6050–6056 (2011).
34. Lamers, C. H. J. et al. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically
retargeted against carbonic anhydrase IX: first clinical experience. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 24, e20–22 (2006).
35. Lamers, C. H. J. et al. Immune responses to transgene and retroviral vector in patients treated with ex vivo-engineered T cells.
Blood 117, 72–82 (2011).
36. Kershaw, M. H. et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin. Cancer
Res. Off. J. Am. Assoc. Cancer Res. 12, 6106–6115 (2006).
37. Abken, H. Adoptive therapy with CAR redirected T cells: the challenges in targeting solid tumors. Immunotherapy 7, 535–544
(2015).
38. Kakarla, S. & Gottschalk, S. CAR T Cells for Solid Tumors: Armed and Ready to Go? Cancer J. 20, 151–155 (2014).
39. Zhang, B.-L. et al. Hurdles of CAR-T cell-based cancer immunotherapy directed against solid tumors. Sci. China Life Sci. (2016).
doi:10.1007/s11427-016-5027-4
40. Chmielewski, M. & Abken, H. TRUCKs: the fourth generation of CARs. Expert Opin. Biol. Ther. 15, 1145–1154 (2015).
41. You, F. et al. Phase 1 clinical trial demonstrated that MUC1 positive metastatic seminal vesicle cancer can be effectively
eradicated by modified Anti-MUC1 chimeric antigen receptor transduced T cells. Sci. China Life Sci. (2016). doi:10.1007/s11427016-5024-7
42. Katz, S. C. et al. Phase I Hepatic Immunotherapy for Metastases Study of Intra-Arterial Chimeric Antigen Receptor-Modified Tcell Therapy for CEA+ Liver Metastases. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 21, 3149–3159 (2015).
43. Brown, C. E. et al. Bioactivity and Safety of IL13Rα2-Redirected Chimeric Antigen Receptor CD8+ T Cells in Patients with
Recurrent Glioblastoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 21, 4062–4072 (2015).
44. John, L. B. et al. Anti-PD-1 Antibody Therapy Potently Enhances the Eradication of Established Tumors By Gene-Modified T
Cells. Clin. Cancer Res. 19, 5636–5646 (2013).
45. Godal, R. et al. Natural Killer Cell Killing of Acute Myelogenous Leukemia and Acute Lymphoblastic Leukemia Blasts by Killer Cell
Immunoglobulin-Like Receptor–Negative Natural Killer Cells after NKG2A and LIR-1 Blockade. Biol. Blood Marrow Transplant. 16,
612–621 (2010).
46. Cooley, S., Burns, L. J., Repka, T. & Miller, J. S. Natural killer cell cytotoxicity of breast cancer targets is enhanced by two distinct
mechanisms of antibody-dependent cellular cytotoxicity against LFA-3 and HER2/neu. Exp. Hematol. 27, 1533–1541 (1999).
47. Gilfillan, S., Ho, E. L., Cella, M., Yokoyama, W. M. & Colonna, M. NKG2D recruits two distinct adapters to trigger NK cell activation
and costimulation. Nat. Immunol. 3, 1150–1155 (2002).
48. Raulet, D. H. Roles of the NKG2D immunoreceptor and its ligands. Nat. Rev. Immunol. 3, 781–790 (2003).
49. Groh, V. et al. Costimulation of CD8alphabeta T cells by NKG2D via engagement by MIC induced on virus-infected cells. Nat.
Immunol. 2, 255–260 (2001).
50. Markiewicz, M. A. et al. Costimulation through NKG2D Enhances Murine CD8+ CTL Function: Similarities and Differences
between NKG2D and CD28 Costimulation. J. Immunol. 175, 2825–2833 (2005).
51. Spear, P., Wu, M.-R., Sentman, M.-L. & Sentman, C. L. NKG2D ligands as therapeutic targets. Cancer Immun. Arch. 13, 8 (2013).
52. Champsaur, M. & Lanier, L. L. Effect of NKG2D ligand expression on host immune responses: Immune response to NKG2D
ligands. Immunol. Rev. 235, 267–285 (2010).
53. Lanier, L. L. NKG2D Receptor and Its Ligands in Host Defense. Cancer Immunol. Res. 3, 575–582 (2015).
54. Raulet, D. H., Gasser, S., Gowen, B. G., Deng, W. & Jung, H. Regulation of Ligands for the NKG2D Activating Receptor. Annu. Rev.
Immunol. 31, 413–441 (2013).
55. Stern-Ginossar, N. & Mandelboim, O. An integrated view of the regulation of NKG2D ligands. Immunology 128, 1–6 (2009).
56. Ljunggren, H.-G. Cancer Immunosurveillance: NKG2D Breaks Cover. Immunity 28, 492–494 (2008).
57. Le Bert, N. & Gasser, S. Advances in NKG2D ligand recognition and responses by NK cells. Immunol. Cell Biol. 92, 230–236 (2014).
58. Pende, D. et al. Major Histocompatibility Complex Class I-related Chain A and UL16-Binding Protein Expression on Tumor Cell
Lines of Different Histotypes Analysis of Tumor Susceptibility to NKG2D-dependent Natural Killer Cell Cytotoxicity. Cancer Res. 62,
6178–6186 (2002).
59. Conejo-Garcia, J. R. et al. Ovarian carcinoma expresses the NKG2D ligand Letal and promotes the survival and expansion of
CD28- antitumor T cells. Cancer Res. 64, 2175–2182 (2004).
60. Barber, A. et al. Chimeric NKG2D Receptor-Bearing T Cells as Immunotherapy for Ovarian Cancer. Cancer Res. 67, 5003–5008
(2007).
61. Li, K. et al. Clinical significance of the NKG2D ligands, MICA/B and ULBP2 in ovarian cancer: high expression of ULBP2 is an
indicator of poor prognosis. Cancer Immunol. Immunother. CII 58, 641–652 (2009).
62. McGilvray, R. W. et al. ULBP2 and RAET1E NKG2D ligands are independent predictors of poor prognosis in ovarian cancer
patients. Int. J. Cancer 127, 1412–1420 (2010).
63. Platonova, S. et al. Profound Coordinated Alterations of Intratumoral NK Cell Phenotype and Function in Lung Carcinoma.
Cancer Res. 71, 5412–5422 (2011).
64. McGilvray, R. W. et al. NKG2D Ligand Expression in Human Colorectal Cancer Reveals Associations with Prognosis and Evidence
for Immunoediting. Clin. Cancer Res. 15, 6993–7002 (2009).
65. Butler, J. E. et al. Proteasome Regulation of ULBP1 Transcription. J. Immunol. 182, 6600–6609 (2009).
66. Maccalli, C. et al. NKG2D engagement of colorectal cancer-specific T cells strengthens TCR-mediated antigen stimulation and
elicits TCR independent anti-tumor activity. Eur. J. Immunol. 33, 2033–2043 (2003).
67. Paschen, A. et al. Differential Clinical Significance of Individual NKG2D Ligands in Melanoma: Soluble ULBP2 as an Indicator of
Poor Prognosis Superior to S100B. Clin. Cancer Res. 15, 5208–5215 (2009).
68. Klöß, S. et al. Increased sMICA and TGFβ 1 levels in HNSCC patients impair NKG2D-dependent functionality of activated NK cells.
OncoImmunology 4, e1055993 (2015).
69. Rebmann, V. et al. Soluble MICA as an independent prognostic factor for the overall survival and progression-free survival of
multiple myeloma patients. Clin. Immunol. 123, 114–120 (2007).
70. Jinushi, M. et al. MHC class I chain-related protein A antibodies and shedding are associated with the progression of multiple
myeloma. Proc. Natl. Acad. Sci. 105, 1285–1290 (2008).
71. Holdenrieder, S. et al. Soluble MICA in malignant diseases. Int. J. Cancer 118, 684–687 (2006).
72. Orozco-Levi, M. et al. Lack of MICA Expression Predicts a Worse Prognosis in Patients with Bladder Cancer. Open J. Pathol. 03,
41–50 (2013).
73. Watson, N. F. S. et al. Expression of the stress-related MHC class I chain-related protein MICA is an indicator of good prognosis
in colorectal cancer patients. Int. J. Cancer J. Int. Cancer 118, 1445–1452 (2006).
74. Dahlberg, C. I. M., Sarhan, D., Chrobok, M., Duru, A. D. & Alici, E. Natural Killer Cell-Based Therapies Targeting Cancer: Possible
Strategies to Gain and Sustain Anti-Tumor Activity. Front. Immunol. 6, (2015).
75. Gras Navarro, A., Björklund, A. T. & Chekenya, M. Therapeutic Potential and Challenges of Natural Killer Cells in Treatment of
Solid Tumors. Front. Immunol. 6, (2015).
76. Morvan, M. G. & Lanier, L. L. NK cells and cancer: you can teach innate cells new tricks. Nat. Rev. Cancer 16, 7–19 (2015).
77. Barber, A., Rynda, A. & Sentman, C. L. Chimeric NKG2D Expressing T Cells Eliminate Immunosuppression and Activate Immunity
within the Ovarian Tumor Microenvironment. J. Immunol. 183, 6939–6947 (2009).
78. Zhang, T. & Sentman, C. L. Cancer Immunotherapy Using a Bispecific NK Receptor Fusion Protein that Engages both T Cells and
Tumor Cells. Cancer Res. 71, 2066–2076 (2011).
79. Nausch, N., Galani, I. E., Schlecker, E. & Cerwenka, A. Mononuclear myeloid-derived ‘suppressor’ cells express RAE-1 and
activate natural killer cells. Blood 112, 4080–4089 (2008).
80. Roy, S. et al. NK Cells Lyse T Regulatory Cells That Expand in Response to an Intracellular Pathogen. J. Immunol. 180, 1729–1736
(2008).
81. Suárez-Álvarez, B., López-Vázquez, A., Baltar, J. M., Ortega, F. & López-Larrea, C. Potential Role of NKG2D and Its Ligands in
Organ Transplantation: New Target for Immunointervention. Am. J. Transplant. 9, 251–257 (2009).
82. Guerra, N. et al. NKG2D-Deficient Mice Are Defective in Tumor Surveillance in Models of Spontaneous Malignancy. Immunity 28,
571–580 (2008).
83. Groh, V., Wu, J., Yee, C. & Spies, T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation.
Nature 419, 734–738 (2002).
84. Ho, V. T. et al. Biologic activity of irradiated, autologous, GM-CSF-secreting leukemia cell vaccines early after allogeneic stem
cell transplantation. Proc. Natl. Acad. Sci. 106, 15825–15830 (2009).
85. Wu, J. Y., Ernstoff, M. S., Hill, J. M., Cole, B. & Meehan, K. R. Ex vivo expansion of non-MHC-restricted cytotoxic effector cells as
adoptive immunotherapy for myeloma. Cytotherapy 8, 141–148 (2006).
86. Meehan, K. R. et al. Adoptive Cellular Therapy using Cells Enriched for NKG2D+CD3+CD8+T Cells after Autologous
Transplantation for Myeloma. Biol. Blood Marrow Transplant. 19, 129–137 (2013).
87. Talebian, L. et al. The natural killer–activating receptor, NKG2D, on CD3+CD8+ T cells plays a critical role in identifying and killing
autologous myeloma cells. Transfusion (Paris) 54, 1515–1521 (2014).
88. Lehner, M. et al. Redirecting T Cells to Ewing’s Sarcoma Family of Tumors by a Chimeric NKG2D Receptor Expressed by
Lentiviral Transduction or mRNA Transfection. PLoS ONE 7, e31210 (2012).
89. Song, D.-G. et al. Chimeric NKG2D CAR-Expressing T Cell-Mediated Attack of Human Ovarian Cancer Is Enhanced by Histone
Deacetylase Inhibition. Hum. Gene Ther. 24, 295–305 (2013).
90. VanSeggelen, H. et al. T Cells Engineered With Chimeric Antigen Receptors Targeting NKG2D Ligands Display Lethal Toxicity in
Mice. Mol. Ther. 23, 1600–1610 (2015).
91. Chang, Y.-H. et al. A Chimeric Receptor with NKG2D Specificity Enhances Natural Killer Cell Activation and Killing of Tumor Cells.
Cancer Res. 73, 1777–1786 (2013).
92. Ruocco, M. G. et al. Suppressing T cell motility induced by anti–CTLA-4 monotherapy improves antitumor effects. J. Clin. Invest.
122, 3718–3730 (2012).
93. Smyth, M. J. et al. NKG2D Recognition and Perforin Effector Function Mediate Effective Cytokine Immunotherapy of Cancer. J.
Exp. Med. 200, 1325–1335 (2004).
94. Gasser, S. The DNA Damage Response Arouses the Immune System. Cancer Res. 66, 3959–3962 (2006).
95. Armeanu, S. et al. Natural killer cell–mediated lysis of hepatoma cells via specific induction of NKG2D ligands by the histone
deacetylase inhibitor sodium valproate. Cancer Res. 65, 6321–6329 (2005).
96. Armeanu, S. et al. Direct and Natural Killer Cell-Mediated Antitumor Effects of Low-Dose Bortezomib in Hepatocellular
Carcinoma. Clin. Cancer Res. 14, 3520–3528 (2008).
97. Zhu, S. et al. The Narrow-Spectrum HDAC Inhibitor Entinostat Enhances NKG2D Expression Without NK Cell Toxicity, Leading
to Enhanced Recognition of Cancer Cells. Pharm. Res. 32, 779–792 (2015).
98. Okita, R. et al. MHC class I chain-related molecule A and B expression is upregulated by cisplatin and associated with good
prognosis in patients with non-small cell lung cancer. Cancer Immunol. Immunother. (2016). doi:10.1007/s00262-016-1814-9
99. Zhao, L., Wang, W.-J., Zhang, J.-N. & Zhang, X.-Y. 5-Fluorouracil and Interleukin-2 Immunochemotherapy Enhances
Immunogenicity of Non-Small Cell Lung Cancer A549 Cells through Upregulation of NKG2D Ligands. Asian Pac. J. Cancer Prev. 15,
4039–4044 (2014).
100. Garrity, D., Call, M. E., Feng, J. & Wucherpfennig, K. W. The activating NKG2D receptor assembles in the membrane with two
signaling dimers into a hexameric structure. Proc. Natl. Acad. Sci. U. S. A. 102, 7641–7646 (2005).
101. Zhang, T., Barber, A. & Sentman, C. L. Generation of Antitumor Responses by Genetic Modification of Primary Human T Cells
with a Chimeric NKG2D Receptor. Cancer Res. 66, 5927–5933 (2006).
102. Barber, A. et al. Chimeric NKG2D receptor–expressing T cells as an immunotherapy for multiple myeloma. Exp. Hematol. 36,
1318–1328 (2008).
103. Zhang, T. & Sentman, C. L. Mouse Tumor Vasculature Expresses NKG2D Ligands and Can Be Targeted by Chimeric NKG2DModified T Cells. J. Immunol. 190, 2455–2463 (2013).
104. Zhang, T., Lemoi, B. A. & Sentman, C. L. Chimeric NK-receptor-bearing T cells mediate antitumor immunotherapy. Blood 106,
1544–1551 (2005).
105. Barber, A., Meehan, K. R. & Sentman, C. L. Treatment of multiple myeloma with adoptively transferred chimeric NKG2D
receptor-expressing T cells. Gene Ther. 18, 509–516 (2011).
106. Zhang, T., Barber, A. & Sentman, C. L. Chimeric NKG2D Modified T Cells Inhibit Systemic T-Cell Lymphoma Growth in a Manner
Involving Multiple Cytokines and Cytotoxic Pathways. Cancer Res. 67, 11029–11036 (2007).
107. Barber, A., Zhang, T. & Sentman, C. L. Immunotherapy with Chimeric NKG2D Receptors Leads to Long-Term Tumor-Free
Survival and Development of Host Antitumor Immunity in Murine Ovarian Cancer. J. Immunol. 180, 72–78 (2008).
108. Spear, P., Barber, A. & Sentman, C. L. Collaboration of chimeric antigen receptor (CAR)-expressing T cells and host T cells for
optimal elimination of established ovarian tumors. OncoImmunology 2, e23564 (2013).
109. Spear, P., Barber, A., Rynda-Apple, A. & Sentman, C. L. NKG2D CAR T-cell therapy inhibits the growth of NKG2D ligand
heterogeneous tumors. Immunol. Cell Biol. 91, 435–440 (2013).
110. Samuels, S. et al. High levels of soluble MICA are significantly related to increased disease-free and disease-specific survival in
patients with cervical adenocarcinoma: sMICA and sHLA-G in cervical cancer. Tissue Antigens 85, 476–483 (2015).
111. Xu, H., Zhu, X.-X. & Chen, J. Abnormal expression levels of sMICA and NKG2D are correlated with poor prognosis in pancreatic
cancer. Ther. Clin. Risk Manag. 11 (2015). doi:10.2147/TCRM.S96869
112. Wang, L.-P. et al. Prognostic significance of serum sMICA levels in non-small cell lung cancer. Eur. Rev. Med. Pharmacol. Sci. 19,
2226–2230 (2015).
113. Dotti, G., Savoldo, B. & Brenner, M. Fifteen years of gene therapy based on chimeric antigen receptors:‘are we nearly there
yet?’ Hum. Gene Ther. 20, 1229–1239 (2009).
114. Morgan, R. A. Human Tumor Xenografts: The Good, the Bad, and the Ugly. Mol. Ther. 20, 882–884 (2012).
115. Barber, A. & Sentman, C. L. Chimeric NKG2D T Cells Require Both T Cell- and Host-Derived Cytokine Secretion and Perforin
Expression to Increase Tumor Antigen Presentation and Systemic Immunity. J. Immunol. 183, 2365–2372 (2009).
116. Spear, P., Barber, A., Rynda-Apple, A. & Sentman, C. L. Chimeric Antigen Receptor T Cells Shape Myeloid Cell Function within
the Tumor Microenvironment through IFN- and GM-CSF. J. Immunol. 188, 6389–6398 (2012).
117. Geissmann, F. et al. Development of Monocytes, Macrophages, and Dendritic Cells. Science 327, 656–661 (2010).
118. Vantourout, P. et al. Immunological Visibility: Posttranscriptional Regulation of Human NKG2D Ligands by the EGF Receptor
Pathway. Sci. Transl. Med. 6, 231ra49–231ra49 (2014).
119. Sotillo, E. et al. Convergence of Acquired Mutations and Alternative Splicing of CD19 Enables Resistance to CART-19
Immunotherapy. Cancer Discov. 5, 1282–1295 (2015).
120. Jackson, H. J. & Brentjens, R. J. Overcoming Antigen Escape with CAR T-cell Therapy. Cancer Discov. 5, 1238–1240 (2015).
121. Wang, L.-C. S. et al. 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, 154–166 (2014).
122. Beatty, G. L. et al. Mesothelin-Specific Chimeric Antigen Receptor mRNA-Engineered T Cells Induce Antitumor Activity in Solid
Malignancies. Cancer Immunol. Res. 2, 112–120 (2014).
123. Stromnes, I. M. et al. T Cells Engineered against a Native Antigen Can Surmount Immunologic and Physical Barriers to Treat
Pancreatic Ductal Adenocarcinoma. Cancer Cell 28, 638–652 (2015).
124. Beatty, G. L. & Moon, E. K. Chimeric antigen receptor T cells are vulnerable to immunosuppressive mechanisms present within
the tumor microenvironment. OncoImmunology 3, e970027 (2014).
125. Moon, E. K. et al. Multifactorial T-cell Hypofunction That Is Reversible Can Limit the Efficacy of Chimeric Antigen ReceptorTransduced Human T cells in Solid Tumors. Clin. Cancer Res. 20, 4262–4273 (2014).
126. Sato, Y. et al. Characterization of the myeloid-derived suppressor cell subset regulated by NK cells in malignant lymphoma.
OncoImmunology 4, e995541 (2015).
127. Khalil, D. N., Smith, E. L., Brentjens, R. J. & Wolchok, J. D. The future of cancer treatment: immunomodulation, CARs and
combination immunotherapy. Nat. Rev. Clin. Oncol. (2016). doi:10.1038/nrclinonc.2016.25
128. Adusumilli, P. S. et al. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4dependent tumor immunity. Sci. Transl. Med. 6, 261ra151–261ra151 (2014).
129. Chekmasova, A. A. et al. 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, 3594–3606 (2010).
130. Davies, D. M. et al. Flexible targeting of ErbB dimers that drive tumorigenesis by using genetically engineered T cells. Mol. Med.
18, 565 (2012).
131. Kong, S. et al. Suppression of Human Glioma Xenografts with Second-Generation IL13R-Specific Chimeric Antigen ReceptorModified T Cells. Clin. Cancer Res. 18, 5949–5960 (2012).
132. Katz, S. C. et al. Regional CAR-T cell infusions for peritoneal carcinomatosis are superior to systemic delivery. Cancer Gene
Ther. (2016). doi:10.1038/cgt.2016.14
133. Spain, L. & Larkin, J. Combination immune checkpoint blockade with ipilimumab and nivolumab in the management of advanced
melanoma. Expert Opin. Biol. Ther. 16, 389–396 (2016).
134. Soriani, A. et al. Chemotherapy-elicited upregulation of NKG2D and DNAM-1 ligands as a therapeutic target in multiple
myeloma. Oncoimmunology 2, e26663 (2013).
135. Vanneman, M. & Dranoff, G. Combining immunotherapy and targeted therapies in cancer treatment. Nat. Rev. Cancer 12, 237–
251 (2012).
136. Liu, C. et al. BRAF Inhibition Increases Tumor Infiltration by T cells and Enhances the Antitumor Activity of Adoptive
Immunotherapy in Mice. Clin. Cancer Res. 19, 393–403 (2013).
137. Frederick, D. T. et al. BRAF Inhibition Is Associated with Enhanced Melanoma Antigen Expression and a More Favorable Tumor
Microenvironment in Patients with Metastatic Melanoma. Clin. Cancer Res. 19, 1225–1231 (2013).
138. Adams, J. L., Smothers, J., Srinivasan, R. & Hoos, A. Big opportunities for small molecules in immuno-oncology. Nat. Rev. Drug
Discov. 14, 603–622 (2015).