Chimeric antigen receptor T-cell therapy - assessment and management of toxicities

Sattva S Neelapu, Sudhakar Tummala, Partow Kebriaei, William Wierda, Cristina Gutierrez, Frederick L Locke, Krishna V Komanduri, Yi Lin, Nitin Jain, Naval Daver, Jason Westin, Alison M Gulbis, Monica E Loghin, John F de Groot, Sherry Adkins, Suzanne E Davis, Katayoun Rezvani, Patrick Hwu, Elizabeth J Shpall, Sattva S Neelapu, Sudhakar Tummala, Partow Kebriaei, William Wierda, Cristina Gutierrez, Frederick L Locke, Krishna V Komanduri, Yi Lin, Nitin Jain, Naval Daver, Jason Westin, Alison M Gulbis, Monica E Loghin, John F de Groot, Sherry Adkins, Suzanne E Davis, Katayoun Rezvani, Patrick Hwu, Elizabeth J Shpall

Abstract

Immunotherapy using T cells genetically engineered to express a chimeric antigen receptor (CAR) is rapidly emerging as a promising new treatment for haematological and non-haematological malignancies. CAR-T-cell therapy can induce rapid and durable clinical responses, but is associated with unique acute toxicities, which can be severe or even fatal. Cytokine-release syndrome (CRS), the most commonly observed toxicity, can range in severity from low-grade constitutional symptoms to a high-grade syndrome associated with life-threatening multiorgan dysfunction; rarely, severe CRS can evolve into fulminant haemophagocytic lymphohistiocytosis (HLH). Neurotoxicity, termed CAR-T-cell-related encephalopathy syndrome (CRES), is the second most-common adverse event, and can occur concurrently with or after CRS. Intensive monitoring and prompt management of toxicities is essential to minimize the morbidity and mortality associated with this potentially curative therapeutic approach; however, algorithms for accurate and consistent grading and management of the toxicities are lacking. To address this unmet need, we formed a CAR-T-cell-therapy-associated TOXicity (CARTOX) Working Group, comprising investigators from multiple institutions and medical disciplines who have experience in treating patients with various CAR-T-cell therapy products. Herein, we describe the multidisciplinary approach adopted at our institutions, and provide recommendations for monitoring, grading, and managing the acute toxicities that can occur in patients treated with CAR-T-cell therapy.

Conflict of interest statement

Competing interests statement

S.S.N. has received research support from Bristol-Myers Squibb, Celgene, Cellectis, Kite Pharma, Merck, and Poseida Therapeutics. S.S.N. has also served as a consultant and/or Scientific Advisory Board member for Celgene, Kite Pharma, Merck, and Novartis. S.T. served as a Scientific Advisory Board member for Kite Pharma. F.L.L. has served as a Scientific Advisory Board member for Kite Pharma, and as a Consultant to Cellular Biomedicine Group. K.V.K. has served as a scientific advisor to and has received research funding from Juno Therapeutics and Kite Pharma. Y.L. has received research funding from Janssen. N.J. has received research support from Abbvie, ADC Therapeutics, Bristol-Myers Squibb, Celgene, Genentech, Incyte, Pharmacyclics, Pfizer, Seattle Genetics, Servier, and Verastem. N.J. has also served on the advisory board and received honorarium from Adaptive Biotechnologies, ADC Therapeutics, Novartis, Novimmune, Pharmacyclics, Pfizer, Servier, and Verastem. N.D. has received research support from Bristol-Myers Squibb, Daichi-Sanky, Incyte, Karyopharm, Pfizer, and Sunesis. N.D. has also received served as a consultant for Incyte, Jazz, Karyopharm, Novartis, Otsuka, Pfizer, and Sunesis. J.W. has received research funding and served on the Advisory Boards for Kite Pharma and Novartis. J.F.d.G. has received research support from Astrazeneca, Deciphera Pharmaceuticals, Eli Lilly, EMD-Serono, Mundipharma, Novartis, Sanofi-Aventis. J.F.d.G. has also served as a consultant or Advisory Board member for AbbVie, Astrazeneca, Celldex, Deciphera Pharmaceuticals, FivePrime Therapeutics, Foundation Medicine, Genentech, Insys Therapeutics, Kadmon, Merck, Novartis, and Novogen. J.F.d.G. is a stock owner of Gilead and Ziopharm Oncology, and his spouse is employed by Ziopharm Oncology. S.A. served as an Advisory Board member for Kite Pharma. K.R. is on the Independent Data Monitoring Committee for Kiadis Pharma. The other authors declare no competing interests.

Figures

Figure 1 |. Clinical case study.
Figure 1 |. Clinical case study.
The findings of key clinical assessments are shown for a representative patient with cytokine-release syndrome and chimeric antigen receptor (CAR)-T-cell-related encephalopathy syndrome after anti-CD19 CAR-T-cell therapy for refractory diffuse large-B-cell lymphoma. a | The graph shows the patient’s maximum temperature (Tmax), maximum heart rate (HRmax), minimum systolic blood pressure (SBPmin), minimum oxygen saturation (O2 satmin), and serum C-reactive protein (CRP) level recorded on each day after anti-CD19 CAR-T-cell therapy. The anti-IL-6 receptor antibody tocilizumab was administered on days 1, 3, and 5 (arrows) for the treatment of hypotension, hypoxia, and encephalopathy, respectively. b | Handwriting samples and mini mental status exam (MMSE) scores obtained on days 4, 5, and 6 after CAR-T-cell therapy; note how the patient’s handwriting was markedly impaired on day 5, despite only a small decrease in their MMSE score. c | 2-[18F]fluoro-2-deoxy-d-glucose PET images showing the retroperitoneal lymph nodes and ileocolic region harbouring lymphoma at baseline (highlighted in red circle; bottom left), and loss of tracer uptake indicative of induction of disease remission at 30 days after infusion of CAR T cells (bottom right).
Figure 2 |. Three-step approach to the…
Figure 2 |. Three-step approach to the assessment and management of acute toxicities associated with chimeric antigen receptor (CAR)-T-cell therapy.
Step 1: the patient’s clinical and biological symptoms should be monitored to determine the nature of the CAR-T-cell-related toxicity, in order to diagnose cytokine-release syndrome (CRS), CAR-T-cell-related encephalopathy syndrome (CRES), and haemophagocytic lymphohistiocytosis/macrophage-activation syndrome (HLH/MAS; BOX 5). Step 2: the severity of CRS, CRES, and HLH/MAS should be graded using the criteria provided in TABLE 2, TABLE 4, and the Common Terminology Criteria for Adverse Events, version 4.03 (CTCAE), respectively. Step 3: the toxicities should be treated according to the management algorithms we have provided for CRS (TABLE 3), CRES (BOX 2), and HLH/MAS (FIG. 3). CARTOX-10, CAR-T-cell-therapy-associated toxicity 10-point neurological assessment; CSF, cerebrospinal fluid; ICP, intracranial pressure.
Figure 3 |. Recommendations for the management…
Figure 3 |. Recommendations for the management of Chimeric antigen receptor (CAR)-T-cell-related haemophagocytic lymphohistiocytosis/macrophage-activation syndrome (HLH/MAS).
HLH/MAS should initially be managed according to the guidelines for cytokine-release syndrome (CRS; TABLE 3), with appropriate subsequent laboratory testing to monitor response to treatment. If the results of these tests reveal no improvement within 48 h, escalation of treatment should be considered.

References

    1. Rosenberg SA & Restifo NP Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348, 62–68 (2015).
    1. June CH, Riddell SR & Schumacher TN Adoptive cellular therapy: a race to the finish line. Sci. Transl Med 7, 280ps7 (2015).
    1. Kochenderfer JN et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 116, 4099–4102 (2010).
    1. Porter DL, Levine BL, Kalos M, Bagg A & June CH Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med 365, 725–733 (2011).
    1. Grupp SA et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N. Engl. J. Med 368, 1509–1518 (2013).
    1. Brentjens RJ et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci. Transl Med 5, 177ra138 (2013).
    1. Cruz CR et al. Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study. Blood 122, 2965–2973 (2013).
    1. Kochenderfer JN et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood 122, 4129–4139 (2013).
    1. Maude SL et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med 371, 1507–1517 (2014).
    1. Davila ML et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci. Transl Med 6, 224ra225 (2014).
    1. Kochenderfer JN et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J. Clin. Oncol 33, 540–549 (2015).
    1. Lee DW 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 385, 517–528 (2015).
    1. Garfall AL et al. Chimeric antigen receptor T cells against CD19 for multiple myeloma. N. Engl. J. Med 373, 1040–1047 (2015).
    1. Fraietta JA et al. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood 127, 1117–1127 (2016).
    1. Brudno JN et al. Allogeneic T cells that express an anti-CD19 chimeric antigen receptor induce remissions of B-cell malignancies that progress after allogeneic hematopoietic stem-cell transplantation without causing graft-versus-host disease. J. Clin. Oncol 34, 1112–1121 (2016).
    1. Kebriaei P et al. Phase I trials using Sleeping Beauty to generate CD19-specific CAR T cells. J. Clin. Invest 126, 3363–3376 (2016).
    1. Turtle CJ et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Invest 126, 2123–2138 (2016).
    1. Turtle CJ et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci. Transl Med 8, 355ra116 (2016).
    1. Turtle CJ et al. Durable molecular remissions in chronic lymphocytic leukemia treated with CD19-specific chimeric antigen receptor-modified T cells after failure of ibrutinib. J. Clin. Oncol 10.1200/JCO.2017.72.8519 (2017).
    1. Kochenderfer JN et al. Lymphoma remissions caused by anti-CD19 Chimeric antigen receptor t cells are associated with high serum interleukin-15 levels. J. Clin. Oncol 35, 1803–1813 (2017).
    1. Hinrichs CS & Rosenberg SA Exploiting the curative potential of adoptive T-cell therapy for cancer. Immunol. Rev 257, 56–71 (2014).
    1. Kochenderfer JN et al. B-Cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 119, 2709–2720 (2012).
    1. Locke FL et al. Phase 1 results of ZUMA-1: a multicenter study of KTE-C19 anti-CD19 CAR T Cell therapy in refractory aggressive lymphoma. Mol. Ther 25, 285–295 (2017).
    1. Neelapu SS et al. KTE-C19 (anti-CD19 CAR T cells) induces complete remissions in patients with refractory diffuse large B-cell lymphoma (DLBCL): results from the pivotal phase 2 ZUMA-1 [abstract]. Blood 128, LBA-6 (2016).
    1. Grupp SA et al. Analysis of a Global registration trial of the efficacy and safety of CTL019 in pediatric and young adults with relapsed/refractory acute lymphoblastic leukemia (ALL) [abstract]. Blood 128, 221 (2016).
    1. Schuster SJ et al. Global pivotal phase 2 trial of the CD19-targeted therapy CTL019 in adult patients with relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL) — an interim analysis [abstract]. Hematol. Oncol 35 (Suppl. S2), 27 (2017).
    1. Neelapu SS et al. Axicabtagene ciloleucel (Axi-cel; KTE-C19) in patients with refractory aggressive non-Hodgkin lymphomas (NHL): primary results of the pivotal trial ZUMA-1 [abstract]. Hematol. Oncol 35 (Suppl. S2), 28 (2017).
    1. Abramson J et al. High CR rates in relapsed/refractory (R/R) aggressive B-NHL treated with the CD19-directed CAR T cell product JCAR017 (TRANSCEND NHL 001) [abstract]. Hematol. Oncol 35 (Suppl. S2), 138 (2017).
    1. Ali SA et al. T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood 128, 1688–1700 (2016).
    1. Lee DW et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 124, 188–195 (2014).
    1. Brudno JN & Kochenderfer JN Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 127, 3321–3330 (2016).
    1. Maude SL, Barrett D, Teachey DT & Grupp SA Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J. 20, 119–122 (2014).
    1. Hu Y et al. Predominant cerebral cytokine release syndrome in CD19-directed chimeric antigen receptor-modified T cell therapy. J. Hematol. Oncol 9, 70 (2016).
    1. Teachey DT et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov. 6, 664–679 (2016).
    1. Ishii K et al. Tocilizumab-refractory cytokine release syndrome (CRS) triggered by chimeric antigen receptor (CAR)-transduced T cells may have distinct cytokine profiles compared to typical CRS. Blood 128, 3358 (2016).
    1. Robbins PF et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J. Clin. Oncol 29, 917–924 (2011).
    1. Koestner W et al. PD-L1 blockade effectively restores strong graft-versus-leukemia effects without graft-versus-host disease after delayed adoptive transfer of T-cell receptor gene-engineered allogeneic CD8+ T cells. Blood 117, 1030–1041 (2011).
    1. Romanski A et al. CD19-CAR engineered NK-92 cells are sufficient to overcome NK cell resistance in B-cell malignancies. J. Cell. Mol. Med 20, 1287–1294 (2016).
    1. Han J et al. CAR-engineered NK cells targeting wild-type EGFR and EGFRvIII enhance killing of glioblastoma and patient-derived glioblastoma stem cells. Sci. Rep 5, 11483 (2015).
    1. Bargou R et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science 321, 974–977 (2008).
    1. Topp MS et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 16, 57–66 (2015).
    1. Teachey DT et al. Cytokine release syndrome after blinatumomab treatment related to abnormal macrophage activation and ameliorated with cytokine-directed therapy. Blood 121, 5154–5157 (2013).
    1. U.S. Department of Health & Human Services. Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0 (2010).
    1. Frey NV et al. Refractory cytokine release syndrome in recipients of chimeric antigen receptor (CAR) T cells. Blood 124, 2296–2296 (2014).
    1. Rose-John S IL-6 trans-signaling via the soluble IL-6 receptor: importance for the pro-inflammatory activities of IL-6. Int. J. Biol. Sci 8, 1237–1247 (2012).
    1. Scheller J, Chalaris A, Schmidt-Arras D & Rose-John S The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim. Biophys. Acta 1813, 878–888 (2011).
    1. Chen F et al. Measuring IL-6 and sIL-6R in serum from patients treated with tocilizumab and/or siltuximab following CAR T cell therapy. J. Immunol. Methods 434, 1–8 (2016).
    1. Singh JA, Beg S & Lopez-Olivo MA Tocilizumab for rheumatoid arthritis. Cochrane Database of Syst. Rev CD008331 (2010).
    1. Deisseroth A et al. FDA approval: siltuximab for the treatment of patients with multicentric Castleman disease. Clin. Cancer Res 21, 950–954 (2015).
    1. Bonifant CL, Jackson HJ, Brentjens RJ & Curran KJ Toxicity and management in CAR T-cell therapy. Mol. Ther. Oncolyt 3, 16011 (2016).
    1. Mihara M et al. Tocilizumab inhibits signal transduction mediated by both mIL-6R and sIL-6R, but not by the receptors of other members of IL-6 cytokine family. Int. Immunopharmacol 5, 1731–1740 (2005).
    1. Zaki MH, Nemeth JA & Trikha M CNTO 328, a monoclonal antibody to IL-6, inhibits human tumor-induced cachexia in nude mice. Int. J. Cancer 111, 592–595 (2004).
    1. Nishimoto N et al. Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease. Blood 112, 3959–3964 (2008).
    1. Paliogianni F, Ahuja SS, Balow JP, Balow JE & Boumpas DT Novel mechanism for inhibition of human T cells by glucocorticoids. Glucocorticoids inhibit signal transduction through IL-2 receptor. J. Immunol 151, 4081–4089 (1993).
    1. Lanza L et al. Prednisone increases apoptosis in in vitro activated human peripheral blood T lymphocytes. Clin. Exp. Immunol 103, 482–490 (1996).
    1. Franchimont D et al. Effects of dexamethasone on the profile of cytokine secretion in human whole blood cell cultures. Regul. Pept 73, 59–65 (1998).
    1. Ozdemir E et al. Cytomegalovirus reactivation following allogeneic stem cell transplantation is associated with the presence of dysfunctional antigen-specific CD8+ T cells. Blood 100, 3690–3697 (2002).
    1. Schultz DR & Arnold PI Properties of four acute phase proteins: C-reactive protein, serum amyloid A protein, α1-acid glycoprotein, and fibrinogen. Semin. Arthritis Rheum 20, 129–147 (1990).
    1. Pepys MB & Hirschfield GM C-Reactive protein: a critical update. J. Clin. Invest 111, 1805–1812 (2003).
    1. Schmidt-Arras D & Rose-John S IL-6 pathway in the liver: from physiopathology to therapy. J. Hepatol 64, 1403–1415 (2016).
    1. Schuster SJ et al. Sustained remissions following chimeric antigen receptor modified T cells directed against CD19 (CTL019) in patients with relapsed or refractory CD19+ lymphomas. Blood 126, 183–183 (2015).
    1. Santomasso B et al. Biomarkers associated with neurotoxicity in adult patients with relapsed or refractory B-ALL (R/R B-ALL) treated with CD19 CAR T cells [abstract]. J. Clin. Oncol 35, (15 Suppl.), 3019 (2017).
    1. Turtle CJ et al. Cytokine release syndrome (CRS) and neurotoxicity (NT) after CD19-specific chimeric antigen receptor- (CAR-) modified T cells [abstract]. J. Clin. Oncol 35, (15 Suppl.), 3020 (2017).
    1. Johnson LA & June CH Driving gene-engineered T cell immunotherapy of cancer. Cell Res. 27, 38–58 (2017).
    1. Sutter R, Semmlack S & Kaplan PW Nonconvulsive status epilepticus in adults — insights into the invisible. Nat. Rev. Neurol 12, 281–293 (2016).
    1. Walker M et al. Nonconvulsive status epilepticus: Epilepsy Research Foundation workshop reports. Epileptic Disord. 7, 253–296 (2005).
    1. Hovinga CA Levetiracetam: a novel antiepileptic drug. Pharmacotherapy 21, 1375–1388 (2001).
    1. Guenther S et al. Chronic valproate or levetiracetam treatment does not influence cytokine levels in humans. Seizure 23, 666–669 (2014).
    1. Reuters. Juno ends development of high-profile leukemia drug after deaths. Reuters (2017).
    1. Harris J Kite reports cerebral edema death in ZUMA-1 CAR T-cell trial. OncLive (2017).
    1. Henter JI et al. HLH-2004: diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis. Pediatr. Blood Cancer 48, 124–131 (2007).
    1. Ramos-Casals M, Brito-Zeron P, Lopez-Guillermo A, Khamashta MA & Bosch X Adult haemophagocytic syndrome. Lancet 383, 1503–1516 (2014).
    1. Jordan MB, Hildeman D, Kappler J & Marrack P An animal model of hemophagocytic lymphohistiocytosis (HLH): CD8+ T cells and interferon gamma are essential for the disorder. Blood 104, 735–743 (2004).
    1. Jordan MB, Allen CE, Weitzman S, Filipovich AH & McClain KL How I treat hemophagocytic lymphohistiocytosis. Blood 118, 4041–4052 (2011).
    1. Tamamyan GN et al. Malignancy-associated hemophagocytic lymphohistiocytosis in adults: Relation to hemophagocytosis, characteristics, and outcomes. Cancer 122, 2857–2866 (2016).
    1. Daver N & Kantarjian H Malignancy-associated haemophagocytic lymphohistiocytosis in adults. Lancet Oncol. 18, 169–171 (2017).
    1. Schram AM & Berliner N How I treat hemophagocytic lymphohistiocytosis in the adult patient. Blood 125, 2908–2914 (2015).
    1. Jordan M et al. A novel targeted approach to the treatment of hemophagocytic lymphohistiocytosis (HLH) with an anti-interferon gamma (IFNγ) monoclonal antibody (mAb), NI-0501: first results from a pilot phase 2 study in children with primary HLH [abstract]. Blood 126, LBA-3 (2015).
    1. Zhou X & Brenner MK Improving the safety of T-Cell therapies using an inducible caspase-9 gene. Exp. Hematol 44, 1013–1019 (2016).
    1. Jackson HJ, Rafiq S & Brentjens RJ Driving CAR T-cells forward. Nat. Rev. Clin. Oncol 13, 370–383 (2016).
    1. Di Stasi A et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med 365, 1673–1683 (2011).
    1. Serafini M et al. Characterization of CD20-transduced T lymphocytes as an alternative suicide gene therapy approach for the treatment of graft-versus-host disease. Hum. Gene Ther 15, 63–76 (2004).
    1. Wang X et al. A transgene-encoded cell surface polypeptide for selection. in vivo tracking, and ablation of engineered cells. Blood 118, 1255–1263 (2011).
    1. Philip B et al. A highly compact epitope-based marker/suicide gene for easier and safer T-cell therapy. Blood 124, 1277–1287 (2014).
    1. Thomis DC et al. A Fas-based suicide switch in human T cells for the treatment of graft-versus-host disease. Blood 97, 1249–1257 (2001).
    1. Sakemura R et al. A Tet-on inducible system for controlling CD19-chimeric antigen receptor expression upon drug administration. Cancer Immunol. Res 4, 658–668 (2016).
    1. Dai H, Wang Y, Lu X & Han W Chimeric Antigen receptors modified T-cells for cancer therapy. J. Natl Cancer Inst 108, djv439 (2016).
    1. Morgan RA 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).
    1. 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).
    1. Parkhurst MR et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol. Ther 19, 620–626 (2011).
    1. Lamers CH 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).
    1. Lamers CH et al. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J. Clin. Oncol 24, e20–e22 (2006).
    1. Maus MV et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol. Res 1, 26–31 (2013).
    1. Brentjens R, Yeh R, Bernal Y, Riviere I & Sadelain M Treatment of chronic lymphocytic leukemia with genetically targeted autologous T cells: case report of an unforeseen adverse event in a phase I clinical trial. Mol. Ther 18, 666–668 (2010).
    1. Chong EA et al. Chimeric antigen receptor modified T cells directed against CD19 (CTL019) in patients with poor prognosis, relapsed or refractory CD19+ follicular lymphoma: prolonged remissions relative to antecedent therapy [abstract]. Blood 128, 1100 (2016).
    1. Locke FL et al. A phase 2 multicenter trial of KTE-C19 (anti-CD19 CAR T Cells) in patients with chemorefractory primary mediastinal B-cell lymphoma (PMBCL) and transformed follicular lymphoma (TFL): interim results from ZUMA-1 [abstract]. Blood 128, 998 (2016).
    1. Russell JA et al. Vasopressin versus norepinephrine infusion in patients with septic shock. N. Engl. J. Med 358, 877–887 (2008).
    1. Frisen L Swelling of the optic nerve head: a staging scheme. J. Neurol. Neurosurg. Psychiatry 45, 13–18 (1982).

Source: PubMed

3
Předplatit