CAR-T Technology and New Opportunities for Tumor Treatment

VYu Pavlova1, ES Livadnyi2

1 SV Belyaev Kemerovo Regional Clinical Hospital, 22 bld. 2 Oktyabrskii pr-t, Kemerovo, Russian Federation, 650066

2 Kemerovo State Medical University, 22a Voroshilova str., Kemerovo, Russian Federation, 650066

For correspondence: Vera Yurevna Pavlova, MD, PhD, 22 bld. 2 Oktyabrskii pr-t, Kemerovo, Russian Federation, 650066; Tel.: +7(951)570-57-86; e-mail: vera.4447.kem@mail.ru

For citation: Pavlova VYu, Livadnyi ES. CAR-T Technology and New Opportunities for Tumor Treatment. Clinical oncohematology. 2021;14(1):149–56. (In Russ).

DOI: 10.21320/2500-2139-2021-14-1-149-156


ABSTRACT

As a cause of death malignant neoplasms come in at the second place after cardiovascular disorders. CAR-T (chimeric antigen receptor of T-cells) therapy is an advanced malignant tumor treatment method. The use of CAR-T lymphocytes refers to adoptive immunotherapy. CAR-T technology is based on “extracting” immune cells (T-lymphocytes) and their genetic modification aimed at acquiring antitumor properties and followed by reinfusion. The advantage of CAR-T therapy in comparison to other treatment methods is that for target cell recognition T-lymphocytes are not dependent on major histocompatibility complex class 1 (MHC-I) molecules. The literature data we collected and analyzed show that this is a fundamentally new and effective treatment method of oncohematological diseases including acute lymphoblastic leukemia, chronic lymphocytic leukemia, and non-Hodgkin’s lymphomas. Clinical trials proved the advantage of CAR-T therapy in comparison to other treatment methods applied in this field. The analysis of literature showed that CAR-T therapy can be reasonably regarded as one of the advanced opportunities for malignant tumor treatment.

Keywords: adoptive immunotherapy, CAR-T lymphocytes, chimeric antigen receptor.

Received: September 20, 2020

Accepted: December 1, 2020

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–Е386. doi: 10.1002/ijc.29210.
  2. Stewart BW, Wilde CP (eds). World cancer report 2014. Lyon: IARC Press, 2014. 619 p.
  3. Plummer M, de Martel C, Vignat J, et al. Global burden of cancers attributable to infections in 2012: a synthetic analysis. Lancet Glob 2016;4(9):e609–e616. doi: 10.1016/S2214-109X(16)30143-7.
  4. Состояние онкологической помощи населению России в 2018 году. Под ред. А.Д. Каприна, В.В. Старинского, Г.В. Петровой. М.: МНИОИ им. П.А. Герцена — филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2019. 236 с.
    [Caprin AD, Starinskii VV, Petrova GV, eds. Sostoyanie onkologicheskoi pomoshchi naseleniyu Rossii v 2018 godu. (The state of cancer care in Russia in 2018.) Moscow: MNIOI im. P.A. Gertsena — a branch of FGBU “NMITs radiologii” Minzdrava Rossii Publ.; 2019. 236 p. (In Russ)]
  5. Здравоохранение в России. Статистический сборник. М.: Росстат, 2011. 326 с.
    [Zdravookhranenie v Rossii. Statisticheskii sbornik. (Health care in Russia. Statistics digest.) Мoscow: Rosstat Publ.; 2011. 326 p. (In Russ)]
  6. Halaleh K, Gale RP. Cancer care in the Palestinian territoried. Lancet Oncol. 2018;19(7):e359–е364. doi: 10.1016/S1470-2045(18)30323-1.
  7. Чикилева И.О., Шубина И.Ж., Киселевский М.В. Влияние регуляторных Т-клеток на функциональную активность натуральных киллеров при иммунотерапии злокачественных опухолей. Вестник РАМН. 2012;67(4):60–4.
    [Chikileva IO, Shubina IZh, Kiselevskii MV. Influence of regulatory T-cells on the functioning of natural killer cells during cancer immunotherapy. Vestnik RAMN. 2012;67(4):60–4. (In Russ)]
  8. Титов К.С., Демидов Л.В., Шубина И.Ж. и др. Технологии клеточной иммунотерапии в лечении больных со злокачественными новообразованиями. Вестник Российского государственного медицинского университета. 2014;1:42–7.
    [Titov KS, Demidov LV, Shubina IZh, et al. Technologies of cell immunotherapy in treatment of cancer patients. Vestnik Rossiiskogo gosudarstvennogo meditsinskogo universiteta; 2014;1:42–7. (In Russ)]
  9. Wahlang B, Falkner KC, Cave MC, et al. Role of Cytochrome P450 Monooxygenase in Carcinogen and Chemotherapeutic Drug Metabolism. Adv Pharmacol. 2015;74:1–33. doi: 10.1016/bs.apha.2015.04.004.
  10. Janeway CA Jr, Travers P, Walport M, et al. Immunobiology: The Immune System in Health and Disease. 5th edition. New York: Garland Science, 2001. 884 p.
  11. Croce CM. Oncogenes and cancer. N Engl J Med. 2008;358(5):502–11. doi: 10.1056/NEJMra072367.
  12. Vicente-Duenas C, Romero-Camarero I, Cobaleda C, et al. Function of oncogenes in cancer development: a changing paradigm. EMBO J. 2013;32(11):1502–13. doi: 10.1038/emboj.2013.97.
  13. Den Haan JM, Arens R, Van Zelm MC. The activation of the adaptive immune system: cross-talk between antigen-presenting cells, T cells and B cells. Immunol Lett. 2014;162(2 Pt B):103–12. doi: 10.1016/j.imlet.2014.10.011.
  14. Emtage PC, Lo AS, Gomes EM, et al. Second-generation anti-carcinoembryonic antigen designer T cells resist activation-induced cell death, proliferate on tumor contact, secrete cytokines, and exhibit superior antitumor activity in vivo: a preclinical evaluation. Clin Cancer Res. 2008;14(24):8112–22. doi: 10.1158/1078-0432.
  15. Maher Immunotherapy of Malignant Disease Using Chimeric Antigen Receptor Engrafted T Cells. ISRN Oncol. 2012;2012:278093. doi: 10.5402/2012/278093.
  16. BonifantL, Jackson HJ, Brentjens RJ, et al. Toxicity and management in CAR T-cell therapy. Mol Ther. 2016;3:16011. doi: 10.1038/mto.2016.11.
  17. Xu J, Wang Q, Xu H, et al. Anti-BCMA CAR-T cells for treatment of plasma cell dyscrasia: case report on POEMS syndrome and multiple myeloma. J Hematol Oncol. 2018;11(1):128. doi: 10.1186/s13045-018-0672-7.
  18. Wang J, Chen S, Xiao W, et al. CAR-T cells targeting CLL-1 as an approach to treat acute myeloid leukemia. J Hematol Oncol. 2018;11(1):7. doi: 10.1186/s13045-017-0553-5.
  19. Wei J, Han X, Bo J, et al. Target selection for CAR-T therapy. J Hematol Oncol. 2019;12(1):62. doi: 10.1186/s13045-019-0758-x.
  20. Si W, Li C, Wei P. Synthetic immunology: T-cell engineering and adoptive immunotherapy. Synth Syst Biotechnol. 2018;3(3):179–85. doi: 10.1016/j.synbio.2018.08.001.
  21. Smith AJ, Oertle J, Warren D, Prato D. Chimeric antigen receptor (CAR) T cell therapy for malignant cancers: Summary and perspective. J Cell Immunother. 2016;2(2):59–68. doi: 10.1016/j.jocit.2016.08.001.
  22. Chmielewski M, Abken H. Trucks: the fourth generation of CARs. Expert Opin Biol Ther. 2015;15(8):1145–54. doi: 10.1517/14712598.2015.1046430.
  23. Zhao Z, Chen Y, Francisco NM, et al. The application of CAR-T cell therapy in hematological malignancies: advantages and challenges. Acta Pharm Sin B. 2018;8(4):539–51. doi: 10.1016/j.apsb.2018.03.001.
  24. Brudno JN, Kochenderfer JN. Recent advances in CAR T-cell toxicity: Mechanisms, manifestations and management. Blood Rev. 2019;34:45–55. doi: 10.1016/j.blre.2018.11.002.
  25. Brudno JN, Maric I, Hartman SD, et al. T cells genetically modified to express an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of poor prognosis relapsed multiple myeloma. J Clin Oncol. 2018;36(22):2267–80. doi: 10.1200/JCO.2018.77.8084.
  26. Maude SL, Frey N, Shaw PA, et al. Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia. N Engl J Med. 2014;371(16):1507–17. doi: 10.1056/NEJMoa1407222.
  27. Fernandez A Cure for Cancer? How CAR T-Cell Therapy is Revolutionizing Oncology. Available from: https://www.labiotech.eu/features/car-t-therapy-cancer-review/ (accessed 25.11.2020).
  28. Porter DL, Hwang WT, Frey NV, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7(303):303ra139. doi: 10.1126/scitranslmed.aac5415.
  29. Schuster SJ, Svoboda J, Chong EA, et al. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N Engl J Med. 2017;377(26):2545–54. doi: 10.1056/NEJMoa1708566.
  30. Pule MA, Savoldo B, Myers GD, et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med. 2008;14(11):1264–70. doi: 10.1038/nm.1882.
  31. Kalos M, Levine BL, Porter DL, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3(95):95ra73. doi: 10.1126/scitranslmed.3002842.
  32. Porter DL, Levine BL, Kalos M, et al. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365(8):725–33. doi: 10.1056/NEJMoa1103849.
  33. Riches JC, Gribben JG. Understanding the immunodeficiency in chronic lymphocytic leukemia: potential clinical implications. Hematol Oncol Clin North Am. 2013;27(2):207–35. doi: 10.1016/j.hoc.2013.01.003.
  34. Ellard R, Stewart O. The EBMT Guidelines for practice. A framework for managing Patient Care, CRS and Neurotoxicity. 1st European CAR T cell meeting, 14–16 February 2019, Paris, France.

Manufacturing of CD19 Specific CAR T-Cells and Evaluation of their Functional Activity in Vitro

AV Petukhov1, VA Markova2, DV Motorin1, AK Titov1, NS Belozerova2, PM Gershovich2, AV Karabel’skii2, RA Ivanov2, EK Zaikova1, EYu Smirnov2, PA Butylin1, AYu Zaritskey1

1 VA Almazov National Medical Research Center, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341

2 Biocad Biotechnology Company, 34-A Svyazi str., Strel’na, Saint Petersburg, Russian Federation, 198515

For correspondence: Andrei Yur’evich Zaritskey, MD PhD, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; Tel.: +7(812)702-68-28, Fax: +7(812)702-37-65; e-mail: zaritskey@gmail.com

For citation: Petukhov AV, Markova VA, Motorin DV, et al. Manufacturing of CD19 Specific CAR T-Cells and Evaluation of their Functional Activity in Vitro. Clinical oncohematology. 2018;11(1):1–9.

DOI: 10.21320/2500-2139-2018-11-1-1-9


ABSTRACT

Background. The most promising variant of adoptive immunotherapy of the B-line oncohematological diseases includes the use of cells with the chimeric antigen receptor (CAR T-cells), that showed extraordinary results in clinical studies.

Aim. To manufacture CAR T-cells for the clinical use and to study their cytotoxicity in vitro.

Methods. Human T-lymphocytes were transduced by the lentiviral vector containing anti-CD19-CAR, RIAD, and GFP genes. The T-cell transduction efficacy was assessed on the basis of GFP protein signal by flow cytometry. Propidium iodide was used to analyse the cell viability. Cytotoxic activity of the manufactured CAR T-cells was studied in the presence of the target cells being directly co-cultivated. Analysis of the number and viability of CAR T-cells and cytokine expression was performed by flow cytometry.

Results. The viability of the transduced T-cells and GFP expression reached 91.87 % and 50.87 % respectively. When cultured in the presence of IL-2 and recombinant CD19 (the target antigen), the amount of CAR-T after 120 h of the process was 1.4 times larger compared with the period of 48 h. In the cytotoxic test of co-cultivation CAR-T with the K562-CD19+ cells the percentage of CAR-T increased to 57 % and 84.5 % after 48 h and 120 h of exposure respectively. When cultured with the K562 cells (test line not expressing CD19) the number of CAR T-cells decreased to 36.2 % within 48 h while the number of K562 cells increased to 58.3 %. The viability of target cells in the experimental and control groups was 3.5 % and 36.74 % respectively. Comparison of IL-6 level in the control and experimental groups revealed that the differences are insignificant, as opposed to the level of other cytokines (IFN-γ, IL-2, TNF) which proved to be different in both groups.

Conclusion. The present work resulted in the production of anti-CD19 CAR T-cells with adequate viability. The in vitro model demonstrated their cytotoxicity. Manufacturing of CAR T-cells for clinical use is the first step of the development of adoptive immunotherapy in the Russian Federation.

Keywords: CAR T-cells, adoptive immunotherapy, acute lymphoblastic leukemia, non-Hodgkin’s lymphomas, lentiviral transduction, graft-versus-host reaction, сytokine release syndrome.

Received: September 15, 2017

Accepted: December 7, 2017

Read in PDF 


REFERENCES

  1. Kanakry CG, Fuchs EJ, Luznik L. Modern approaches to HLA-haploidentical blood or marrow transplantation. Nat Rev Clin Oncol. 2015;13(1):10–24. doi: 10.1038/nrclinonc.2015.128.
  2. Podhorecka M, Markowicz J, Szymczyk A, Pawlowski J. Target therapy in hematological malignances: new monoclonal antibodies. Int Sch Res Not. 2014;2014(3):1–16. doi: 10.1155/2014/701493.
  3. Hussaini M. Biomarkers in Hematological Malignancies: A Review of Molecular Testing in Hematopathology. Cancer Control. 2015;22(2):158–66. doi: 10.1177/107327481502200206.
  4. Forman SJ, Rowe JM. The myth of the second remission of acute leukemia in the adult. Blood. 2013;121(7):1077–82. doi: 10.1182/blood-2012-08-234492.
  5. Im A, Pavletic SZ. Immunotherapy in hematologic malignancies: past, present, and future. J Hematol Oncol. 2017;10(1):94. doi: 10.1186/s13045-017-0453-8.
  6. Luskin MR, DeAngelo DJ. Chimeric Antigen Receptor Therapy in Acute Lymphoblastic Leukemia Clinical Practice. Curr Hematol Malig Rep. 2017;12(4):370–9. doi: 10.1007/s11899-017-0394-x.
  7. Fesnak A, June CH, Levine BL. Engineered T cells: the promise and challenges of cancer immunotherapy. Nat Rev Cancer. 2016;16(9):566–81. doi: 10.1038/nrc.2016.97.
  8. Lim W, June C. The Principles of Engineering Immune Cells to Treat Cancer. Cell. 2017;168(4):724–40. doi: 10.1016/j.cell.2017.01.016.
  9. Sadelain M, Brentjens R, Riviere I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013;3(4):388–98. doi: 10.1158/2159-8290.CD-12-0548.
  10. Brentjens RJ, Davila ML, Riviere I, et al. CD19-Targeted T Cells Rapidly Induce Molecular Remissions in Adults with Chemotherapy-Refractory Acute Lymphoblastic Leukemia. Sci Transl Med. 2013;5(177):177ra38. doi: 10.1126/scitranslmed.3005930.
  11. Maude SL, Frey N, Shaw P, et al. Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia. N Engl J Med. 2014;371(16):1507–17. doi: 10.1056/NEJMoa1407222.
  12. Turtle CJ, Hanafi L-A, Berger C, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest. 2016;126(6):2123–38. doi: 10.1172/JCI85309.
  13. Lee DW, Kochenderfer JN, Stetler-Stevenson M, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: A phase 1 dose-escalation trial. Lancet. 2015;385(9967):517–28. doi: 10.1016/S0140-6736(14)61403-3.
  14. Onea AS, Jazirehi AR. CD19 chimeric antigen receptor (CD19 CAR)-redirected adoptive T-cell immunotherapy for the treatment of relapsed or refractory B-cell Non-Hodgkin’s Lymphomas. Am J Cancer Res. 2016;6(2):403–24.
  15. Kebriaei P, Singh H, Huls MH, et al. Phase I trials using Sleeping Beauty to generate CD19-specific CAR T cells. J Clin Invest. 2016;126(9):3363–76. doi: 10.1172/JCI86721.
  16. ICML 2017: Data From the TRANSCEND Trial of JCAR017 in Relapsed and Refractory Aggressive B-Cell Non-Hodgkin Lymphoma — The ASCO Post. Available from: http://www.ascopost.com/News/57764 (accessed 7.10.2017).
  17. Locke FL, Neelapu SS, Bartlett NL, et al. Abstract CT019: Primary results from ZUMA-1: a pivotal trial of axicabtagene ciloleucel (axicel; KTE-C19) in patients with refractory aggressive non-Hodgkin lymphoma (NHL). Cancer Res. 2017;77(13 Suppl):CT019. doi: 10.1158/1538-7445.AM2017-CT019.
  18. Kalos M, Levine BL, Porter DL, et al. T Cells with Chimeric Antigen Receptors Have Potent Antitumor Effects and Can Establish Memory in Patients with Advanced Leukemia. Sci Transl Med. 2011;3(95):95ra73. doi: 10.1126/scitranslmed.3002842.
  19. Porter DL, Hwang W-T, Frey NV, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7(303):303ra139. doi: 10.1126/scitranslmed.aac5415.
  20. Jensen MC, Riddell SR. Designing chimeric antigen receptors to effectively and safely target tumors. Curr Opin Immunol. 2015;33:9–15. doi: 10.1016/j.coi.2015.01.002.
  21. Павлова А.А., Масчан М.А., Пономарев В.Б. Адоптивная иммунотерапия генетически модифицированными Т-лимфоцитами, экспрессирующими химерные антигенные рецепторы. Онкогематология. 2017;12(1):17–32. doi: 10.17650/1818-8346-2017-12-1-17-32. [Pavlova AА, Maschan MА, Ponomarev VB. Adoptitive immunotherapy with genetically engineered T lymphocytes modified to express chimeric antigen receptors. Oncohematology. 2017;12(1):17–32. doi: 10.17650/1818-8346-2017-12-1-17-32. (In Russ)]
  22. Dai H, Wang Y, Lu X, Han W. Chimeric Antigen Receptors Modified T-Cells for Cancer Therapy. J Natl Cancer Inst. 2016;108(7):1–15. doi: 10.1093/jnci/djv439.
  23. Abate-Daga D, Davila ML. CAR models: next-generation CAR modifications for enhanced T-cell function. Mol Ther Oncolytics. 2016;3:16014. doi: 10.1038/mto.2016.14.
  24. Holzinger A, Barden M, Abken H. The growing world of CAR T cell trials: a systematic review. Cancer Immunol Immunother. 2016;65(12):1433–50. doi: 10.1007/s00262-016-1895-5.
  25. Jensen MC, Popplewell L, Cooper LJ, et al. Antitransgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19-specific chimeric antigen receptor redirected T cells in humans. Biol Blood Marrow Transplant. 2010;16(9):1245–56. doi: 10.1016/j.bbmt.2010.03.014.
  26. Gong MC, Latouche JB, Krause A, et al. Cancer patient T cells genetically targeted to prostate-specific membrane antigen specifically lyse prostate cancer cells and release cytokines in response to prostate-specific membrane antigen. Neoplasia. 1999;1(2):123–7.
  27. Davila ML, Sadelain M. Biology and clinical application of CAR T cells for B cell malignancies. Int J Hematol. 2016;104(1):6–17. doi: 10.1007/s12185-016-2039-6.
  28. Park JH, Geyer MB, Brentjens RJ. CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date. Blood. 2016;127(26):3312–20. doi: 10.1182/blood-2016-02-629063.
  29. Grupp S, Kalos M, Barrett D, et al. Chimeric Antigen Receptor–Modified T Cells for Acute Lymphoid Leukemia. N Engl J Med. 2013;368(16):1509–18. doi: 10.1056/NEJMoa1215134.
  30. Yu H, Sotillo E, Harrington C, et al. Repeated loss of target surface antigen after immunotherapy in primary mediastinal large B cell lymphoma. Am J Hematol. 2017;92(1):E11–E13. doi: 10.1002/ajh.24594.
  31. Sotillo E, Barrett DM, Black KL, et al. Convergence of Acquired Mutations and Alternative Splicing of CD19 Enables Resistance to CART-19 Immunotherapy. Cancer Discov. 2015;5(12):1282–95. doi: 10.1158/2159-8290.CD-15-1020.
  32. Fischer J, Paret C, El Malki K, et al. CD19 Isoforms Enabling Resistance to CART-19 Immunotherapy Are Expressed in B-ALL Patients at Initial Diagnosis. J Immunother. 2017;40(5):187–95. doi: 10.1097/CJI.0000000000000169.
  33. Jacoby E, Nguyen SM, Fountaine TJ, et al. CD19 CAR immune pressure induces B-precursor acute lymphoblastic leukaemia lineage switch exposing inherent leukaemic plasticity. Nat Commun. 2016;7:12320. doi: 10.1038/ncomms12320.
  34. Gardner R, Wu D, Cherian S, et al. Acquisition of a CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy. Blood. 2016;127(20):2406–10. doi: 10.1182/blood-2015-08-665547.
  35. Zah E, Lin M-Y, Silva-Benedict A, et al. T Cells Expressing CD19/CD20 Bispecific Chimeric Antigen Receptors Prevent Antigen Escape by Malignant B Cells. Cancer Immunol Res. 2016;4(6):498–508. doi: 10.1158/2326-6066.CIR-15-0231.
  36. Shah NN, Stetler-Stevenson M, Yuan CM, et al. Minimal Residual Disease Negative Complete Remissions Following Anti-CD22 Chimeric Antigen Receptor (CAR) in Children and Young Adults with Relapsed/Refractory Acute Lymphoblastic Leukemia (ALL). Blood. 2016;128(22):650.
  37. Davila ML, Riviere I, Wang X, et al. Efficacy and Toxicity Management of 19-28z CAR T Cell Therapy in B Cell Acute Lymphoblastic Leukemia. Sci Transl Med. 2014;6(224):224ra25. doi: 10.1126/scitranslmed.3008226.
  38. Long AH, Haso WM, Shern JF, et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med. 2015;21(6):581–90. doi: 10.1038/nm.3838.
  39. Hay KA, Turtle CJ. Chimeric Antigen Receptor (CAR) T Cells: Lessons Learned from Targeting of CD19 in B-Cell Malignancies. Drugs. 2017;77(3):237–45. doi: 10.1007/s40265-017-0690-8.
  40. Wehbi VL, Tasken K. Molecular mechanisms for cAMP-mediated immunoregulation in T cells – role of anchored protein kinase a signaling units. Front Immunol. 2016;7:1–19. doi: 10.3389/fimmu.2016.00222.
  41. Newick K, O’Brien S, Sun J, et al. Augmentation of CAR T-cell Trafficking and Antitumor Efficacy by Blocking Protein Kinase A Localization. Cancer Immunol Res. 2016;4(6):541–51. doi: 10.1158/2326-6066.CIR-15-0263.
  42. Sanjana NE, Shalem O, Zhang F. Improved vectors and genome-wide libraries for CRISPR screening. Nat Methods. 2014;11(8):783–4. doi: 10.1038/nmeth.3047.
  43. Kochenderfer JN, Feldman SA, Zhao Y, et al. Construction and Preclinical Evaluation of an Anti-CD19 Chimeric Antigen Receptor. J Immunother. 2009;32(7):689–702. doi: 10.1097/CJI.0b013e3181ac6138.
  44. Wang K, Wei G, Liu D. CD19: a biomarker for B cell development, lymphoma diagnosis and therapy. Exp Hematol Oncol. 2012;1(1):36. doi: 10.1186/2162-3619-1-36.
  45. Uckun FFM, Jaszcz W, Ambrus JJL, et al. Detailed Studies on Expression and Function of CD19 Surface Determinant by Using B43 Monoclonal Antibody and the Clinical Potential of Anti-CD19 Immunotoxins. Blood. 1988;71(1):13–29.
  46. Wei G, Ding L, Wang J, et al. Advances of CD19-directed chimeric antigen receptor-modified T cells in refractory/relapsed acute lymphoblastic leukemia. Exp Hematol Oncol. 2017;6(1):10. doi: 10.1186/s40164-017-0070-9.
  47. Barrett DM, Singh N, Hofmann TJ, et al. Interleukin 6 Is Not Made By Chimeric Antigen Receptor T Cells and Does Not Impact Their Function. Blood. 2016;128(22):2016–7.
  48. Singh N, Hofmann TJ, Gershenson Z, et al. Monocyte lineage-derived IL-6 does not affect chimeric antigen receptor T-cell function. Cytotherapy. 2017;19(7):867–80. doi: 10.1016/j.jcyt.2017.04.001.
  49. Hartmann J, Schussler‐Lenz M, Bondanza A, Buchholz CJ. Clinical development of CAR T cells—challenges and opportunities in translating innovative treatment concepts. EMBO Mol Med. 2017;9(9):1183–97. doi: 10.15252/emmm.201607485.
  50. Hagen T. Novartis Sets a Price of $475,000 for CAR T-Cell Therapy. Available from: http://www.onclive.com/web-exclusives/novartis-sets-a-price-of-475000-for-car-tcell-therapy (accessed 31.10.2017).
  51. Yang Y, Jacoby E, Fry TJ. Challenges and opportunities of allogeneic donor-derived CAR T cells. Curr Opin Hematol. 2015;22(6):509–15. doi: 10.1097/MOH.0000000000000181.
  52. Li H, Zhao Y. Increasing the safety and efficacy of chimeric antigen receptor T cell therapy. Protein Cell. 2017;8(8):573–89. doi: 10.1007/s13238-017-0411-9.