Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Aug 3.
Published in final edited form as: Cancer J. 2014 Mar-Apr;20(2):119–122. doi: 10.1097/PPO.0000000000000035

Managing Cytokine Release Syndrome Associated With Novel T Cell-Engaging Therapies

Shannon L Maude *, David Barrett *, David T Teachey *, Stephan A Grupp *,
PMCID: PMC4119809  NIHMSID: NIHMS607703  PMID: 24667956

Abstract

Chimeric antigen receptor (CAR)-modified T cells and bispecific T cell–engaging antibodies have demonstrated dramatic clinical responses in recent clinical trials. The hallmark of these novel highly active immunotherapies is nonphysiologic T cell activation, which has correlated not only with greatly increased efficacy but also with notable toxicity in some cases. We and others have observed a cytokine release syndrome (CRS), which correlates with both toxicity and efficacy in patients receiving T cell–engaging therapies. In addition to elevations in effector cytokines, such as interferon-γ, cytokines associated with hemophagocytic lymphohistiocytosis or macrophage activation syndrome, such as interleukin (IL)-10 and IL-6, may also be markedly elevated. Whereas corticosteroids may control some of these toxicities, their potential to block T cell activation and abrogate clinical benefit is a concern. Detailed studies of T cell proliferation and the resultant immune activation produced by these novel therapies have led to more targeted approaches that have the potential to provide superior toxicity control without compromising efficacy. One approach we have developed targets IL-6, a prominent cytokine in CRS, using the IL-6R antagonist tocilizumab. We will review the pathophysiology and management options for CRS associated with T cell–engaging therapies.

Keywords: Acute lymphoblastic leukemia, chimeric antigen receptor, blinatumomab, cytokine release syndrome


T cell–engaging therapies have shown remarkable promise for highly refractory B-cell malignancies; however, this type of immunotherapy presents unique challenges in toxicity management. By linking target antigen-expressing malignant cells to cytotoxic T cells, T cell–engaging therapies harness the cell-mediated immune response and direct it against cancerous cells, bypassing the major histocompatibility complex. T cells engineered to express a chimeric antigen receptor (CAR) with CD19 specificity and blinatumomab, a bispecific antibody linking CD3+ T cells with CD19+ cells, are 2 such therapies that have shown dramatic responses in early-phase clinical trials for relapsed/refractory B-cell leukemias and lymphomas.15 These trials have demonstrated high complete response (CR) rates; however, CRs have been associated with cytokine release syndrome (CRS), which ranges from mild to life threatening.

Our group has shown that some patients develop clinical and laboratory findings similar to patients with macrophage activation syndrome/hemophagocytic lymphohistiocytosis (MAS/HLH) and that cytokine-directed therapy can abrogate life-threatening CRS symptoms without compromising efficacy.2,6 This review will focus on the clinical manifestations and management of CRS associated with novel T cell–engaging therapies.

CART-19/CTL019 CLINICAL ACTIVITY

Chimeric antigen receptor–modified T cells (CART) use adoptive transfer of T lymphocytes engineered to express a single-chain fragment variable region (scFv) domain linked to the signaling domain of the T cell receptor (TCR),7 plus various costimulatory domains such as CD28 and CD137 (4-1BB).1,812 For CART-19, the scFv is directed against CD19, an attractive target for most B-cell malignancies, owing to its near-universal expression on malignant cells and specificity for the B-cell lineage. CD19 is expressed throughout B-cell development from the early pro-B stage through mature B cells.13 Whereas the expression of chimeric antibodies in T cells was first described by Eshhar et al14 more than 20 years ago, poor expansion and persistence of the engineered T cells limited clinical efficacy for many years. However, we and others have shown that CART therapy has promising activity in both chronic lymphocytic leukemia (CLL) and acute lymphoblastic leukemia (ALL). Porter et al were the first to report that CART-19 was effective in adults with refractory CLL. In 3 patients with high disease burdens, CART-19 therapy produced sustained CRs in 2 patients and a partial response in the third.1 Our group also reported striking efficacy in relapsed pediatric B-ALL.2 As recently reported at the ASH meeting (December 2013), we have treated 22 pediatric patients with relapsed/refractory ALL with CART-19 with a CR rate of 86% (19/22) at day 28. Sustained remissions of 2 to 20 months have been achieved in 12 of 17 evaluable patients with a median follow-up of 3.6 months. More than 70% of these patients had relapsed after allogeneic stem cell transplant, and none experience graft versus host disease despite a median donor T cell chimerism of 100%.15 Other groups also have reported striking efficacy of CAR-modified T cells in relapsed/refractory CLL and ALL.8,10,16

BLINATUMOMAB CLINICAL ACTIVITY

Blinatumomab, a member of a new class of bispecific antibodies, links CD19-expressing target cells with T effector cells.17 Resultant cytotoxic T cell engagement leads to target cell lysis. Owing to its short half-life, blinatumomab is given by continuous intravenous infusion in 4-week cycles. In the first clinical trial of blinatumomab, efficacy was demonstrated in adults with refractory lymphomas, with a 35% objective response rate.3 In a phase 2 study of adults with minimal residual disease–positive ALL, 16 of 21 patients became negative for minimal residual disease.4 Of the 11 patients who did not subsequently receive allogeneic hematopoietic stem cell transplant, 6 patients remain in remission with a median follow-up of almost 3 years.18 Early data from a phase 2 dose escalation trial in adults with refractory/relapsed ALL suggest a 75% CR rate in patients with significant disease burden at the start of treatment.4 Finally, early data from a phase 1 dose-escalation trial of blinatumomab in children with relapsed/refractory B-ALL (NCT01471782) demonstrate a 47% CR rate,19 and a phase 2 trial for relapsed/refractory pediatric ALL opened in October 2013.

CYTOKINE RELEASE SYNDROME AND HLH AFTER T CELL–ENGAGING THERAPIES

The most common toxicity associated with the novel immunotherapies CART-19 and blinatumomab is CRS, a constellation of inflammatory symptoms resulting from cytokine elevations associated with T cell engagement and proliferation. In most patients, CRS symptoms are mild and flulike, with fevers and myalgias. However, some patients experience a severe inflammatory syndrome, including vascular leak, hypotension, pulmonary edema, and coagulopathy, resulting in multiorgan system failure; and recently, a CRS-related death after blinatumomab was reported.19 Cytokine elevations are measurable in most patients, but the degree of elevation may not correlate with severity of CRS or response to therapy. Moreover, some patients experience symptoms without marked cytokine elevation, whereas others demonstrate laboratory findings out of proportion to clinical symptoms.20

The management of CRS can be challenging. Fulminant CRS may be life threatening; however, some degree of cytokine release is likely a necessary consequence of T cell activation and therefore efficacy. Corticosteroids are an obvious choice to blunt CRS owing to their known efficacy in disorders that involve activated T cells, such as graft versus host disease and inflammatory processes.21 Indeed, corticosteroids have been incorporated into the routine supportive care for blinatumomab therapy. However, the potential negative impact on therapeutic T cell proliferation remains a concern for cellular therapies. In the original report of CART-19 efficacy, Porter et al1 observed a partial response in a patient who received corticosteroids early after CART-19 infusion. Yet, our group and Brentjens et al2,10 have since used short-term steroids to treat life-threatening CRS in some cases without compromising CART proliferation or efficacy. In vitro, steroids reduce cytokine levels without affecting the T cell activation produced by bispecific T cell engagers.22 However, the potential to blunt efficacy with corticosteroid use remains a concern, which needs to be balanced with their well-documented benefit in disorders of excess inflammation.

Another strategy to manage CRS is to target elevated cytokines directly. The most significantly elevated cytokines in the CRS associated with CART-191 and blinatumomab20 are IL-10, IL-6, and IFN-γ. IL-10, primarily produced by monocytes/macrophages, regulates both innate and cell-mediated immunity by inhibiting activated macrophages.23 IL-10 can also be produced by mast cells, B cells, regulatory T cells, and helper T cells (TH2), but not commonly by cytotoxic T lymphocytes. As a negative regulator, IL-10 may not be an ideal cytokine to target in CRS. Conversely, IL-6 is an inflammatory cytokine involved in a large number of processes, both within the immune system, such as neutrophil trafficking, acute phase response, angiogenesis, B cell differentiation, and autoantibody production, and outside it, such as bone and lipid metabolism.24 IL-6 is produced by monocytes/macrophages, dendritic cells, T cells, fibroblasts, keratinocytes, endothelial cells, adipocytes, myocytes, mesangial cells, and osteoblasts. Interferon-γ, also an inflammatory cytokine, is involved in macrophage activation, TH1 differentiation, major histocompatibility complex class 1 induction, and B-cell isotype switching; and it is produced by cytotoxic T cells, helper T cells (TH1), and natural killer cells.25 As an effector cytokine released by activated cytotoxic T cells after engagement, IFN-γ elevations are both anticipated and likely required for efficacy, making it a potentially undesirable target for toxicity management. Whereas it is not known if IL-6 plays a role in T cell proliferation through a feedback loop, it is thought to be primarily involved in the inflammatory process.

In patients with severe CRS associated with T cell–engaging therapies, IL-6 levels peak during maximal T cell proliferation. We theorized that targeting IL-6 might reduce CRS toxicity without compromising clinical efficacy of T cell–engaging therapies. In our experience, IL-6 blockade by tocilizumab has resulted in rapid, dramatic reversal of life-threatening CRS in patients treated with CART-19 or blinatumomab.2,6 Tocilizumab, a recombinant humanized monoclonal antibody against the IL-6 receptor, blocks IL-6 from binding to its receptor, both in membrane-bound and soluble states.26,27 With a primary indication for juvenile idiopathic arthritis (JIA), tocilizumab is approved by the Food and Drug Administration for children as young as 2 years. It is also approved for adults with rheumatoid arthritis and for Castleman disease in Japan. Tocilizumab has been extensively studied in adults, with 8 randomized controlled trials treating more than 2000 patients,26,27 and in children in phase 1 to phase 3 trials for JIA.2830 Tocilizumab was safely combined with other immunosuppressant medications in most of these trials, with the most common adverse events being liver enzyme elevations and cytopenias. Although grade 3 or greater transaminase elevations (>5× the upper limit of normal) are rare (italic>1%), some degree of liver enzyme elevation occurred in 34% to 41% of patients in the tocilizumab arm compared to 17% in the placebo arm; however, many of these studies combined tocilizumab with hepatotoxic agents, including methotrexate. Cytopenias, including neutropenia and thrombocytopenia, were reported with repeat dosing; however, less than 1% of patients developed severe neutropenia (bold>500/μL) or thrombo-cytopenia (<50,000/μL). Whereas tocilizumab is typically dosed every 2 to 4 weeks, extended treatment is not necessary in the management of CRS, which is self-limited.

In a subset of patients with severe CRS, we have observed a symptom pattern similar to that seen in HLH or MAS. In these patients, high fevers have been accompanied by hepatosplenomegaly, liver dysfunction, coagulopathy, hypofibrinogenemia, and profound hyperferritinemia. Furthermore, the cytokine profile observed in patients treated with CART-19 or blinatumomab mirrors that seen in HLH.3134 In addition to elevations in IL-10, IL-6, and IFN-γ, elevated IL-2R, MCP-1, and MIP1B are reported in patients treated with CART-19 and blinatumomab, with normal levels of IL-1B, IL-4, IL-5, IL-7, IL-12, IL-13, IL-17, tumor necrosis factor (TNF)-α, and granulocyte macrophage colony-stimulating factor (GM-CSF).6,20,35 Whereas engagement of cytotoxic T cells would be expected to produce high levels of IFN-γ, elevations of IL-6 and IL-10 are better explained by HLH/MAS. Individual cytokine levels can vary markedly between patients; however, the similarity of the patterns observed in patients receiving CART-19 or blinatumomab and the patients with HLH is quite striking. HLH is a rare disorder of immune regulation leading to pathologic immune activation and excessive inflammation. It is characterized by prolonged fever, splenomegaly, cytopenias, hyperferritinemia, hypertriglyceridemia, coagulopathy, decreased natural killer cell function, and elevated cytokines.36 Primary HLH is an inherited disorder caused by defective cytolytic granule exocytosis. In patients with homozygous mutations in one of several genes, leading to loss of function of proteins involved in cytolytic granule exocytosis, HLH can present in infancy with minimal or no trigger. MAS is a form of secondary HLH triggered by immune-activating processes, such as infection, malignancy, or autoimmune diseases. Although MAS was initially thought not to be inherited, new data suggest that many patients with secondary HLH have heterozygous mutations in the same genes that are mutated in primary HLH, leading to mild dysfunctions in the same immune regulatory processes.3639 In these patients, HLH presents in childhood or adulthood after a significant immune-activating trigger. As seen later in the text, T cell–engaging therapies can provide such a significant trigger.

T cell–engaging therapies, like CART-19 and blinatumomab, cause transient but profound T cell proliferation and cytokine release, which could trigger a cascade of immune activation sufficient to cause HLH/MAS, both in predisposed patients and patients with no known mutations in HLH-causing genes. The parallels in both clinical and laboratory findings between CRS and HLH/MAS suggest similar pathophysiology. In fact, CRS associated with T cell therapies may represent a spectrum of symptoms, with a subset of patients developing significant, if temporary, HLH/MAS. Disease burden, degree of T cell proliferation, or genetic predisposition may increase the risk of developing HLH/MAS in some patients. Further studies may find merit in screening for HLH-predisposing mutations in high-risk genes, such as PRF1, MUNC13-4, STXBP2, and STX11 to predict patients at higher risk of developing HLH/MAS. Moreover, we propose prospective monitoring for HLH/MAS in patients treated with CART therapies and bispecific T cell–engaging antibodies and have incorporated this monitoring in our practice. HLH/MAS can develop whether T cells are activated through the native TCR, as is the case with blinatumomab, or a CAR that bypasses the TCR; therefore, this monitoring may be warranted in other cellular therapies as well.

Although tocilizumab has rapidly reversed life-threatening manifestations of CRS caused by CART-19 or blinatumomab, it should be noted that there is some concern that tocilizumab should be avoided if MAS is suspected. This concern stems from a case report suggesting that tocilizumab may temporarily mask or control clinical symptoms of MAS in patients with JIA, thereby delaying definitive therapy.40 In MAS caused by an auto-immune disease, symptoms may flare when IL-6 blockade is lifted if definitive therapy is not initiated; however, this is less of a concern with the transient MAS associated with T cell therapies.

Other cytokine-directed approaches to managing CRS could be considered. Inhibitors of MCP-1 and MIP1B are in development, but not in clinical use, whereas inhibitors of IL-2R, IL-1R, and TNF-α have been used clinically. Tumor necrosis factor α is elevated in inflammatory syndromes but does not seem to be elevated after T cell therapy. Tumor necrosis factor α can be targeted by etanercept, which has demonstrated efficacy in rheumatologic disorders41 but showed no obvious clinical benefit in severe CRS after CART-19 therapy in a single pediatric patient with ALL.2 Soluble IL-2 receptor (CD25) is elevated after blinatumomab or CART-19 treatment in some patients and is markedly elevated in patients with HLH. Daclizumab, a monoclonal antibody against CD25, has potential efficacy in HLH;42,43 however, it is no longer commercially available, and targeting CD25, which is present on activated T cells, may compromise efficacy of the cell therapy. Elevated IL-1β is prominent in JIA, and anakinra, a recombinant form of the IL1 receptor antagonist (IL1Ra), has been used for HLH/MAS associated with JIA.44 Whereas marked elevations in IL-1β have not been observed in our few patient observations to date after blinatumomab or CART-19 therapy, a subset of patients do have modest increases in IL-1α, raising the possibility that anakinra could have a role in managing CRS after T cell–engaging therapies.

In summary, there are a number of potential treatment options for CRS associated with T cell–engaging therapies; however, many of these carry the theoretical risk of inhibiting T cell activity and could impair treatment efficacy. Our data suggest that tocilizumab is effective at reversing CRS without inhibiting the efficacy of CART19 or blinatumomab.

CONCLUSION

As we enter the era of highly active T cell–engaging therapies, as exemplified by the bispecific T cell–engaging antibody blinatumomab and CAR-modified T cell therapies such as CART-19/CTL019, it has become apparent that the high degrees of T cell activation that result in dramatic clinical responses are accompanied by significant toxicities. Cytokine release syndrome is a potentially life-threatening complication of the nonphysiologic T cell activation that is a hallmark of T cell–engaging therapies. We have now demonstrated that nonphysiologic T cell activation can produce abnormal macrophage activation, mimicking HLH, a syndrome that may contribute significantly to these toxicities. The challenge in toxicity management is controlling symptoms without compromising efficacy. In the case of CAR-modified T cell therapies, significant clinical responses may require a high degree of in vivo proliferation, and it is the ability to achieve this proliferation that has resulted in both powerful efficacy results and increased toxicity. Targeted therapies may control CRS while maintaining efficacy. Our data suggest that IL-6 blockade by tocilizumab is effective at reversing CRS and controlling HLH/MAS without inhibiting the efficacy of CART-19 or blinatumomab. As T cell–engaging therapies show great promise, further studies are needed to determine optimal toxicity management.

Acknowledgments

This work was supported in part by grants from the National Institutes of Health (R01CA116660), the Leukemia and Lymphoma Society, Pennsylvania Department of Health, Cookies for Kids Cancer, Solving Kids Cancer, the St. Baldrick’s Foundation, the WW Smith Charitable Trust, and the Weinberg Funds.

Footnotes

S.L.M. has served on the advisory board for Incyte Corporation. S.A.G. has provided research, consulting, and clinical trial support to Novartis. The remaining authors have disclosed that they have no significant relationships with, or financial interest in, any commercial companies pertaining to this article.

References

  • 1.Porter DL, Levine BL, Kalos M, et al. Chimeric antigen receptor–modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365:725–733. doi: 10.1056/NEJMoa1103849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Grupp SA, Kalos M, Barrett D, et al. Chimeric antigen receptor–modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368:1509–1518. doi: 10.1056/NEJMoa1215134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Bargou R, Leo E, Zugmaier G, et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science. 2008;321:974–977. doi: 10.1126/science.1158545. [DOI] [PubMed] [Google Scholar]
  • 4.Topp MS, Goekbuget N, Zugmaier G, et al. Anti-CD19 BiTE blinatumomab induces high complete remission rates in adult patients with relapsed B-precursor ALL: updated results of an on-going phase II trial. Blood: ASH Annual Meeting Abstracts. 2011;118:252. [Google Scholar]
  • 5.Topp MS, Kufer P, Gokbuget N, et al. Targeted therapy with the T-cell–engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol. 2011;29:2493–2498. doi: 10.1200/JCO.2010.32.7270. [DOI] [PubMed] [Google Scholar]
  • 6.Teachey DT, Rheingold SR, Maude SL, et al. Cytokine release syndrome after blinatumomab treatment related to abnormal macrophage activation and ameliorated with cytokine directed therapy. Blood. 2013;121:5154–5157. doi: 10.1182/blood-2013-02-485623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lee DW, Barrett DM, Mackall C, et al. The future is now: chimeric antigen receptors as new targeted therapies for childhood cancer. Clin Cancer Res. 2012;18:2780–2790. doi: 10.1158/1078-0432.CCR-11-1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Brentjens RJ, Riviere I, Park JH, et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011;118:4817–4828. doi: 10.1182/blood-2011-04-348540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Kochenderfer JN, Wilson WH, Janik JE, 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. 2010;116:4099–4102. doi: 10.1182/blood-2010-04-281931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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;5:177ra38. doi: 10.1126/scitranslmed.3005930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Sadelain M, Brentjens R, Riviere I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013;3:388–398. doi: 10.1158/2159-8290.CD-12-0548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Milone MC, Fish JD, Carpenito C, et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther. 2009;17:1453–1464. doi: 10.1038/mt.2009.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Portell CA, Advani AS. Antibody therapy for acute lymphoblastic leukemia. Curr Hematol Malig Rep. 2012;7:153–159. doi: 10.1007/s11899-012-0120-7. [DOI] [PubMed] [Google Scholar]
  • 14.Eshhar Z, Waks T, Gross G, et al. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci U S A. 1993;90:720–724. doi: 10.1073/pnas.90.2.720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Grupp SA, Frey NV, Aplenc R, et al. T cells engineered with a chimeric antigen receptor (CAR) targeting CD19 (CTL019) produce significant in vivo proliferation, complete responses and long-term persistence without GVHD in children and adults with relapsed, refractory ALL. Blood. 2013;122:67. [Google Scholar]
  • 16.Kochenderfer JN, Dudley ME, Carpenter RO, et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood. 2013;122:4129–4139. doi: 10.1182/blood-2013-08-519413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wolf E, Hofmeister R, Kufer P, et al. BiTEs: bispecific antibody constructs with unique anti-tumor activity. Drug Discov Today. 2005;10:1237–1244. doi: 10.1016/S1359-6446(05)03554-3. [DOI] [PubMed] [Google Scholar]
  • 18.Topp MS, Gokbuget N, Zugmaier G, et al. Long-term follow-up of hematologic relapse-free survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL. Blood. 2012;120:5185–5187. doi: 10.1182/blood-2012-07-441030. [DOI] [PubMed] [Google Scholar]
  • 19.Gore L, Zugmaier G, Handgretinger R, et al. Cytological and molecular remissions with blinatumomab treatment in second or later bone marrow relapse in pediatric acute lymphoblastic leukemia (ALL) J Clin Oncol. 2013;31(suppl):abstr 7020. [Google Scholar]
  • 20.Klinger M, Brandl C, Zugmaier G, et al. Immunopharmacologic response of patients with B-lineage acute lymphoblastic leukemia to continuous infusion of T cell-engaging CD19/CD3-bispecific BiTE antibody blinatumomab. Blood. 2012;119:6226–6233. doi: 10.1182/blood-2012-01-400515. [DOI] [PubMed] [Google Scholar]
  • 21.Martin PJ, Rizzo JD, Wingard JR, et al. First- and second-line systemic treatment of acute graft-versus-host disease: recommendations of the American Society of Blood and Marrow Transplantation. Biol Blood Marrow Transplant. 2012;18:1150–1163. doi: 10.1016/j.bbmt.2012.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Brandl C, Haas C, d’Argouges S, et al. The effect of dexamethasone on polyclonal T cell activation and redirected target cell lysis as induced by a CD19/CD3-bispecific single-chain antibody construct. Cancer Immunol Immunother. 2007;56:1551–1563. doi: 10.1007/s00262-007-0298-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Iyer SS, Cheng G. Role of interleukin 10 transcriptional regulation in inflammation and autoimmune disease. Crit Rev Immunol. 2012;32:23–63. doi: 10.1615/critrevimmunol.v32.i1.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Mihara M, Hashizume M, Yoshida H, et al. IL-6/IL-6 receptor system and its role in physiological and pathological conditions. Clin Sci (Lond) 2012;122:143–159. doi: 10.1042/CS20110340. [DOI] [PubMed] [Google Scholar]
  • 25.Saha B, Jyothi Prasanna S, Chandrasekar B, et al. Gene modulation and immunoregulatory roles of interferon gamma. Cytokine. 2010;50:1–14. doi: 10.1016/j.cyto.2009.11.021. [DOI] [PubMed] [Google Scholar]
  • 26.Singh JA, Beg S, Lopez-Olivo MA. Tocilizumab for rheumatoid arthritis: a Cochrane systematic review. J Rheumatol. 2011;38:10–20. doi: 10.3899/jrheum.100717. [DOI] [PubMed] [Google Scholar]
  • 27.Singh JA, Wells GA, Christensen R, et al. Adverse effects of biologics: a network meta-analysis and Cochrane overview. Cochrane Database Syst Rev. 2011:CD008794. doi: 10.1002/14651858.CD008794.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Woo P, Wilkinson N, Prieur AM, et al. Open label phase II trial of single, ascending doses of MRA in Caucasian children with severe systemic juvenile idiopathic arthritis: proof of principle of the efficacy of IL-6 receptor blockade in this type of arthritis and demonstration of prolonged clinical improvement. Arthritis Res Ther. 2005;7:R1281–R1288. doi: 10.1186/ar1826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Yokota S, Miyamae T, Imagawa T, et al. Therapeutic efficacy of humanized recombinant anti-interleukin-6 receptor antibody in children with systemic-onset juvenile idiopathic arthritis. Arthritis Rheum. 2005;52:818–825. doi: 10.1002/art.20944. [DOI] [PubMed] [Google Scholar]
  • 30.De Benedetti F, Brunner HI, Ruperto N, et al. Randomized trial of tocilizumab in systemic juvenile idiopathic arthritis. N Engl J Med. 2012;367:2385–2395. doi: 10.1056/NEJMoa1112802. [DOI] [PubMed] [Google Scholar]
  • 31.Tang Y, Xu X, Song H, et al. Early diagnostic and prognostic significance of a specific Th1/Th2 cytokine pattern in children with haemophagocytic syndrome. British journal of haematology. 2008;143:84–91. doi: 10.1111/j.1365-2141.2008.07298.x. [DOI] [PubMed] [Google Scholar]
  • 32.Xu XJ, Tang YM, Song H, et al. Diagnostic accuracy of a specific cytokine pattern in hemophagocytic lymphohistiocytosis in children. J Pediatr. 2012;160:984–990. e1. doi: 10.1016/j.jpeds.2011.11.046. [DOI] [PubMed] [Google Scholar]
  • 33.Olin RL, Nichols KE, Naghashpour M, et al. Successful use of the anti-CD25 antibody daclizumab in an adult patient with hemophagocytic lymphohistiocytosis. Am J Hematol. 2008;83:747–749. doi: 10.1002/ajh.21236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Tamura K, Kanazawa T, Tsukada S, et al. Increased serum monocyte chemoattractant protein-1, macrophage inflammatory protein-1beta, and interleukin-8 concentrations in hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 2008;51:662–668. doi: 10.1002/pbc.21660. [DOI] [PubMed] [Google Scholar]
  • 35.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;3:95ra73. doi: 10.1126/scitranslmed.3002842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Risma K, Jordan MB. Hemophagocytic lymphohistiocytosis: updates and evolving concepts. Curr Opin Pediatr. 2012;24:9–15. doi: 10.1097/MOP.0b013e32834ec9c1. [DOI] [PubMed] [Google Scholar]
  • 37.Zhizhuo H, Junmei X, Yuelin S, et al. Screening the PRF1, UNC13D, STX11, SH2D1A, XIAP, and ITK gene mutations in Chinese children with Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 2012;58:410–414. doi: 10.1002/pbc.23216. [DOI] [PubMed] [Google Scholar]
  • 38.Zhang K, Jordan MB, Marsh RA, et al. Hypomorphic mutations in PRF1, MUNC13-4, and STXBP2 are associated with adult-onset familial HLH. Blood. 2011;118:5794–5798. doi: 10.1182/blood-2011-07-370148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Trizzino A, zur Stadt U, Ueda I, et al. Genotype-phenotype study of familial haemophagocytic lymphohistiocytosis due to perforin mutations. J Med Genet. 2008;45:15–21. doi: 10.1136/jmg.2007.052670. [DOI] [PubMed] [Google Scholar]
  • 40.Shimizu M, Nakagishi Y, Kasai K, et al. Tocilizumab masks the clinical symptoms of systemic juvenile idiopathic arthritis-associated macrophage activation syndrome: the diagnostic significance of interleukin-18 and interleukin-6. Cytokine. 2012;58:287–294. doi: 10.1016/j.cyto.2012.02.006. [DOI] [PubMed] [Google Scholar]
  • 41.Horneff G, Schmeling H, Biedermann T, et al. The German etanercept registry for treatment of juvenile idiopathic arthritis. Ann Rheum Dis. 2004;63:1638–1644. doi: 10.1136/ard.2003.014886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Tomaske M, Amon O, Bosk A, et al. Alpha-CD25 antibody treatment in a child with hemophagocytic lymphohistiocytosis. Med Pediatr Oncol. 2002;38:141–142. doi: 10.1002/mpo.1294. [DOI] [PubMed] [Google Scholar]
  • 43.Lackner H, Urban C, Sovinz P, et al. Hemophagocytic lymphohistiocytosis as severe adverse event of antineoplastic treatment in children. Haematologica. 2008;93:291–294. doi: 10.3324/haematol.11704. [DOI] [PubMed] [Google Scholar]
  • 44.Verbsky JW, White AJ. Effective use of the recombinant interleukin 1 receptor antagonist anakinra in therapy resistant systemic onset juvenile rheumatoid arthritis. J Rheumatol. 2004;31:2071–2075. [PubMed] [Google Scholar]

RESOURCES