
Chimeric antigen receptor T cell immunotherapy‑associated hemophagocytic lymphohistiocytosis: Pathogenesis, clinical manifestation, diagnosis and management compared with cytokine release syndrome (Review)
- Authors:
- Published online on: May 26, 2025 https://doi.org/10.3892/mmr.2025.13578
- Article Number: 213
-
Copyright: © Hu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Chimeric antigen receptor (CAR) T cell therapy serves an important role in cancer treatment (1) and CAR T cells have been approved for treating hematological malignancy (2). CAR T cells may cause lethal toxicity through several mechanisms (3–5) and cytokine release syndrome (CRS) is the most common toxic effect (6). Its symptoms often mimic those of influenza, including fever, headache, malaise, myalgia, rigor and anorexia (1). However, some severe cases progress to life-threatening symptoms, such as hypotension, hypoxia, pulmonary edema, pleural effusion and tachycardia (7). CAR T cell-associated hemophagocytic lymphohistiocytosis (CARHLH), which carries a high risk of lethality, shares similar clinical manifestations with CRS, such as fever, multiorgan dysfunction, hyperferritinemia and hypercytokinemia (8,9).
A retrospective analysis by the European Society for Blood and Marrow Transplantation (EBMT) included 201 patients receiving CAR-T therapy from 114 centers across 24 countries, with a CARHLH incidence rate of 3.48% between 2016 and 2018 (10). A retrospective study of 59 patients with relapsed/refractory B cell acute lymphoblastic leukemia/lymphoblastic lymphoma treated with CD22 CAR T cells reported that 52 patients (88.1%) developed CRS, of whom 21 (40.4%) developed CARHLH (all following CRS onset), including six of 12 patients with severe (grade3/4) CRS (11). Another study on 27 patients with relapsed and/or refractory CD19-positive acute lymphoblastic leukemia treated with CD19 CAR T cells reported that 11 (40%) had CRS and four (14.8%) had CRS and CARHLH (12); all patients with CARHLH either concurrently or previously experienced CRS. Typically, CRS resolves or is fully resolved prior to the onset of CARHLH (11,13). The progression of CRS and CARHLH is connected, making it challenging to distinguish between these (11,13).
However, previous studies have indicated that the pathogenesis, treatment and prognosis of these diseases differ (11,13,14). Patients suspected of having CARHLH should be diagnosed as soon as possible and promptly receive effective treatment to control disease progression and improve outcomes (1,5,10,11,15–18). Therefore, early identification and prompt treatment are essential for improving patient prognosis.
HLH
HLH is a severe inflammatory syndrome caused by abnormal immune regulation (19). A study of patients with primary HLH (pHLH) revealed that the outcome is usually rapidly fatal, with a median survival <2 months without timely treatment (20). HLH is characterized by uncontrolled activation of T cells and macrophages (21). The cardinal features include hyperferritinemia, fever, hepatosplenomegaly, cytopenia and hemophagocytic phenomena in the spleen, liver, bone marrow and lymph nodes (17,22,23). HLH is classified as primary or secondary, based on its etiology. pHLH arises from immune deficiencies associated with mutations in genes responsible for the cytotoxic functions of natural killer (NK) cells and cytotoxic T lymphocytes (CTLs). The onset of pHLH commonly occurs in infants aged <1 year (21). Secondary HLH (sHLH) is more common in adults with underlying conditions including infection, autoimmune disease, autoinflammatory responses and cancer (22,24). Infection is the most frequent trigger of sHLH (25).
Genetic or acquired immunoregulatory defects imbalance the regulatory pathways responsible for the natural termination of immune or inflammatory responses, leading to sustained and excessive activation of T and antigen-presenting cells. Abnormal NK cell function is also observed in patients with HLH (19). NK cells regulate the initial responses of antigen-presenting cells to pathogens and attenuate signals communicated by antigen-driven T cells. NK cells also serve a role in the contraction of immune responses by culling activated T cells and histiocytes during the later stages of antigen-driven activation (26). The abnormal function of CTLs and NK cells may lead to continuous activation and proliferation of macrophages triggered by antigens. Activated macrophages persistently proliferate and release cytokines that promote the recruitment and expansion of lymphocytes and other inflammatory cells. This can lead to organ damage and systemic manifestations of hypercytokinemia. In addition, activated macrophages non-selectively phagocytose hematopoietic components, resulting in observable hemophagocytosis in the bone marrow, lymph nodes and spleen (27).
HLH is a rare condition: The incidence of HLH (including pHLH and sHLH) in individuals aged <18 years in the United States is 1/100,000 (28). In Sweden, the incidence of familial HLH (fHLH) in children is 1.2/1,000,000 (20), whereas that of sHLH in children is 0.19/100,000 (29). HLH is a life-threatening disease. A Swedish study of 32 children with fHLH, with a median survival of only 2–3 months (20). However, mortality rates differ between subtypes, with malignancy-associated HLH having the worst prognosis (17,18). The 2-month survival rate for malignancy-associated HLH was 52% overall from 1997 to 2018 in Sweden (29).
CAR T cell therapy
CAR T cell therapy is based on the isolation of T cells from peripheral blood, which are genetically engineered to express CARs. These cells are expanded in vitro and then re-infused into patients with cancer. CAR T cells undergo non-specific expansion in vivo and simulate immune responses against cancer cells (30). This modification enables the modified T cells to recognize and attack tumor cells independently of major histocompatibility complex (MHC) engagement (2,31–33). CAR T cell therapy targets tumor cells independently of antigen processing and presentation, making them resistant to tumor escape mechanisms associated with MHC loss. Furthermore, this approach can effectively recognize antigens such as glycolipids, proteins with abnormal glycosylation and conformational epitopes (34,35). CAR T cell therapy has notable benefits in the treatment of various malignancies (33,36–38). The response rates of patients with relapsed or refractory B non-Hodgkin's lymphoma is 52–82% (38,39). In patients with relapsed or refractory B cell ALL, the rate of complete remission is 90%, with a 2-year overall survival rate of 73% and an event-free survival rate of 49.5% (40). The objective and complete remission rates in patients with relapsed or refractory multiple myeloma are 85 and 45%, respectively. The median progression-free survival is 11.8 months (41).
However, CAR T cells may damage normal tissue by cross-reacting with antigens that are not expressed in tumor cells (3,4). CAR-targeted tumor-associated antigens may be expressed in normal tissue, leading to CAR T cell-mediated damage to normal tissue cells (5). CAR T cells can cause complications such as CRS and neurotoxicity. Additionally, both the malignancies and the chemotherapy that patients may receive before CAR T cell infusion can impair immune function, thereby increasing the risk of infection (42). CAR T cell therapy has the potential to trigger life-threatening toxicity, such as CARHLH (5).
CRS
CRS is the most frequent adverse effect associated with CAR T cell therapy. A meta-analysis of 2,592 patients from 84 studies who received CAR T cell therapy reported that the all-grade CRS rate is 77%, while the grade ≥3 CRS rate is 29% (43). This is a systemic inflammatory response triggered by the release of pro-inflammatory cytokines from activated CAR T, ruptured cancer and other immune cells. The interplay between CAR T and cancer cells triggers the activation of immune cells, such as macrophages and endothelial cells, which amplifies the production of cytokines (such as IL-6, IFN-γ, IL-8 and IL-10), leading to a cytokine storm (44).
Furthermore, cytokines, perforin and granzyme from CAR T cells induce pyroptosis in tumor cells. Pyroptosis is a form of programmed cell death distinct from apoptosis, marked by cell swelling, lysis and the release of cellular contents and proinflammatory factors (Fig. 1) (45). The majority of CRS cases develop within the first 14 days of infusion and persist for 7 or 8 days (7,39,46,47). In most cases, CRS resolves naturally without intervention, and patients typically require only symptomatic care (1,5,7,15,16,48,49). The initial symptoms include fever, headache, malaise, myalgias, and rigors (1). However, some severe cases progress to life-threatening symptoms, such as hypotension, hypoxia, pulmonary edema, pleural effusion and tachycardia (7). These symptoms may lead to multiorgan failure and require intensive care management (1,7). A meta-analysis by Lei et al (43) reported a CRS mortality rate <1%.
CARHLH
CARHLH is a severe toxicity associated with CAR T cell therapy (9). CARHLH typically manifests with symptoms such as fever, hepatosplenomegaly, peripheral blood trilineage or bilineage cytopenia, elevated cytokine levels and increased soluble CD25. CARHLH can be life-threatening and is associated with multiorgan dysfunction, infection and coagulopathy (9). A case report described a patient with multiple myeloma who achieved a stringent complete response in the primary disease evaluation before CAR T cell infusion (50). However, after the infusion of CAR T cells, the patient developed CARHLH and succumbed to multiple infections caused by bacteria, fungi and viruses (50). The cause of sHLH varies, with numerous underlying factors, and its pathophysiological mechanisms are incompletely understood. The pathogenesis of CARHLH, a subtype of sHLH associated with immunotherapy, has not been fully elucidated. Lichtenstein et al (11) observed an elevated T:NK cell ratio and a decreased percentage of peripheral blood monocytes in patients with CARHLH. Patients who develop CARHLH exhibit relative NK cell lymphopenia before infusion, which becomes more severe during peak CAR expansion in patients with CARHLH (11).
NK cells inhibit excessive activation and expansion of CD8+ T cells (51). Deficiency of NK cells may lead to the over-expansion of CAR T cells and delayed T cell contraction (51). CAR T and ruptured tumor cells release proinflammatory cytokines in HLH. Previous studies have demonstrated that tumor cell pyroptosis results in the release of IL-18 and other cytokines (52,53). Excessive IL-18 can initiate signaling cascades that activate nuclear factor κB or STAT3, MAPK and JNK pathways, ultimately leading to the robust production and dissemination of IFN-γ (52,53). This activates macrophages to release more cytokines, thereby creating a positive feedback loop of inflammation, and generating the clinical manifestations associated with CARHLH. Moreover, excessive IL-6 production during the disease may affect the synthesis of perforin and granzyme, leading to impaired NK cell activity and perpetuating immune imbalance (Fig. 2) (49,54,55).
To the best of our knowledge, there is a lack of large studies investigating CARHLH (19). CARHLH is rapidly becoming a fatal condition. In a clinical study (56), 100 patients with relapsed or refractory large B cell lymphoma received CAR T cell therapy. CARHLH occurred in five cases, of which two were treated with anakinra (a recombinant human IL-1 receptor antagonist), etoposide and corticosteroids but succumbed to CARHLH on days 15 and 71 of treatment (56). A multicenter clinical trial (38) of 101 patients treated with CD19 CAR T cell therapy observed that 94 patients (93%) developed CRS, and one patient developed CRS progressed and succumbed to CARHLH. By contrast, the CRS manifestations in the other patients resolved after treatment, with one patient succumbing to cardiac arrest (38). A retrospective review (57) of 105 patients with relapsed or refractory large B cell lymphoma who received CAR T cell therapy reported that six patients (5.7%) developed CARHLH. The progression-free and overall survival rates of patients who developed CARHLH were markedly worse than those of patients without HLH (57). Hines et al (12) conducted a cohort study of 27 patients with relapsed or refractory CD19 acute lymphoblastic leukemia. Following CAR T cell therapy, four patients (14.8%) developed CARHLH, one succumbed to CARHLH and three succumbed to leukemia, with a median survival of 44.5 days. The aforementioned study revealed that patients who developed CARHLH experienced worse outcomes, a greater incidence of non-response to CAR T cell therapy and markedly decreased overall survival (12).
Although CARHLH and CRS have similar clinical manifestations, there are differences in their pathogenesis. Patients with CARHLH have more severe and persistent clinical manifestations; therefore, early recognition and rational treatment are key (11).
Diagnosis and differentiation of CRS and CARHLH
Diagnosis of CRS is primarily based on clinical symptoms. The American Society for Transplantation and Cellular Therapy (ASTCT) defines CRS following immunotherapy as ‘a supraphysiologic response following any immune therapy that results in the activation or engagement of endogenous or infused T cells and/or other immune effector cells. Symptoms can be progressive, must include fever at the onset, and may include hypotension, capillary leak (hypoxia) and end organ dysfunction’ (58). CRS should be suspected if ≥1 of the following four manifestations occurs within 3 weeks of CAR T cell infusion: Fever (temperature ≥38°C), hypotension (systolic blood pressure <90 mmHg), hypoxia (arterial oxygen saturation <90%) or organ toxicity (48). Laboratory tests are key for identifying other diseases, such as sepsis and tumor lysis syndrome, because the clinical manifestations of CRS are non-specific. If CRS is suspected, a thorough evaluation of the patient should be conducted, and treatment should be initiated accordingly (1). The ASTCT consensus provides grading criteria for CRS based on clinical manifestation (Table I) (58).
To the best of our knowledge, there are currently no standardized diagnostic criteria for CARHLH, although both the guidelines released by the ASTCT in 2023 and the previous HLH diagnostic criteria are used in conjunction with the clinical manifestations of patients to diagnose CARHLH (50,59,60). The diagnosis should be accompanied by close clinical observation and repeated laboratory tests. The HLH-2004 criteria, H-score, Takagi criteria and MD Anderson criteria are used to help diagnose CARHLH (Table II) (9,10,48,61–63). The 2023 ASTCT expert recommendation on CARHLH/IEC-HS use the term ‘immune effector cell-associated HLH-like syndrome (IEC-HS)’ to describe CARHLH and describe the definition of IEC-HS. The guideline recommends a diagnosis of CARHLH based on clinical manifestations, with a considerable elevation in serum ferritin levels being the primary criterion for diagnosis (Table II) (9). ASTCT proposed a grading schema for this condition (Table III). The MD Anderson criteria (10) are the diagnostic standards for CARHLH. However, their clinical application has not yet become widespread (10). The HLH-2004 criteria are currently the accepted diagnostic protocol for HLH and are used to diagnose CARHLH (10,61). The H-score identifies patients with HLH by assigning points based on clinical manifestation (62), whereas the Takagi criteria diagnose HLH based on a combination of pathological and clinical characteristics (63). The HLH-2004 protocol, H-score and Takagi criteria were not developed specifically for CARHLH, and therefore have limitations in diagnosing CARHLH. A study (14) of 20 adult and 16 pediatric HLH patients from Belgium. The study found that the H-score demonstrated better sensitivity and specificity than the HLH-2004 protocol in the early diagnosis of the disease (14). For diagnosing HLH in adults, the H-score is more effective than the traditional HLH-2004 criteria. However, the sensitivity and specificity of the H-score decrease in the diagnosis of deteriorating diseases (14).
Early recognition of CARHLH in patients with CRS and the timely initiation of effective treatment are critical for patient prognosis. The ASTCT expert recommendations state that patients with HLH-like toxicities experience either a preceding or concurrent CRS and suggest the use of markedly elevated ferritin levels as the primary criterion for diagnosing CARHLH (9). Ferritin is the most frequently used laboratory test to distinguish CRS and CARHLH; however, the threshold value is not yet standardized (9,64). Elevated ferritin levels may indicate CARHLH; however, low ferritin levels have a negative predictive value (10). Zu et al (44) retrospectively studied 99 patients treated with B cell maturation antigen (BCMA). Serum parameter monitoring revealed elevated ferritin, aspartate aminotransferase and lactate dehydrogenase (LDH) levels in patients with CARHLH and CRS, as well as a marked peak difference between the two groups; elevated parameters in patients with CARHLH persisted for an extended duration (44). Similar patterns were observed during monitoring of cytokines. Compared with patients with CRS, the levels of IFN-γ, granzyme B, IL-1a, IL-1b, IL-10, IL-12, IL-33 and chemokine are markedly increased in patients with CARHLH (11,44).
Numerous studies (11,44) have attempted to explore the predictors of CARHLH. Lichtenstein et al (11) developed a predictive model of CD22 CAR T cells using IFN-γ and baseline myeloid T:NK cell ratios on day 2 post-CRS to predict the probability that patients with CRS develop CARHLH in the future. Zu et al (44) also developed a CARHLH prediction model for BCMA therapy that used fibrinogen levels on day 3 post-CRS and LDH levels on day 3 post-CRS to predict CARHLH in patients with CRS. Further clinical studies are required to clarify the predictive effects of these models, which are still in the research stage and have not been extensively applied in clinical practice.
Treatment of CRS and CARHLH
According to MD Anderson Cancer Center, patients should be hospitalized and closely monitored for ≥7 days following CAR T cell infusion (48). The treatment protocol should be formulated based on the overall clinical condition of the patient (15). Management of ASTCT grade 1 CRS primarily involves supportive measures including rehydration, antipyretics, oxygenation and elevation of blood pressure. The option of anti-cytokine treatment may be evaluated in patients with moderate-to-severe CRS (15,48,58). IL-6 is a key factor in the pathogenesis of CRS, and its expression is associated with the severity of CRS following CAR T cell therapy. Furthermore, anti-IL-6 therapy does not affect the expansion or efficacy of CAR T cells (38,48,65,66). Consequently, anti-IL-6 is the preferred treatment option for patients with severe CRS (48,66). Numerous case reports demonstrate the efficacy of tocilizumab (an IL-6 monoclonal antibody) in managing CAR T cell-associated CRS (50,67,68). The Society for Immunotherapy of Cancer (SITC) and American Society of Clinical Oncology (ASCO) (69,70) recommend the use of anti-cytokine therapy and corticosteroids in CRS (Table IV). Compared with SITC, ASCO is more proactive in the use of anti-cytokine therapy and corticosteroids (69,70).
The current first-line treatment regimen for HLH consists of etoposide, cyclosporine A and dexamethasone, as well as intrathecal methotrexate and dexamethasone and is indicated for most types of HLH. Due to the toxic side effects of chemotherapy, maximum supportive treatment should be provided during the treatment period. Bone marrow transplantation is the sole curative option for treating pHLH, malignancy-associated HLH and Epstein-Barr virus-related HLH (19). Disease remission prior to transplantation is key for patient prognosis (71). Biological agent-targeted therapies are a promising treatment option: In a clinical study of 34 patients with pHLH (27 treated and seven primary), 8-week treatment with emapalumab (an IFN-γ monoclonal antibody) had effective rates of 63 and 65%, respectively (72). Ge et al (73) included 21 patients with pHLH (12 treated and nine primary) using a ruxolitinib (a JAK inhibitor)-based regimen, with a 1-year overall survival rate of 90.5%; all patients achieved objective remission within the first 8 weeks, with 19 (90.5%) achieving complete remission.
The treatment of CARHLH has not yet been standardized. The ASTCT CRS guidelines published in 2019 (58) suggest that there is no need to distinguish CARHLH from CRS. However, there is an increasing consensus that the early identification of CARHLH and prompt treatment are essential for improving patient outcomes (58,69,70). A case report described two patients with CARHLH: A patient who received timely diagnosis and treatment experienced improvement in CARHLH after therapy. By contrast, a patient who did not receive prompt medical attention, quickly succumbed from multi-organ failure (68). The MD Anderson Cancer Center recommends patients with suspected CARHLH initially be considered as having CRS and should be treated with corticosteroids and anti-IL-6 therapy (48).
A retrospective study by the EBMT reported that the most frequently used combination regimen for the treatment of CARHLH is corticosteroids combined with chemotherapy, followed by corticosteroids combined with chemotherapy and monoclonal antibodies or anti-cytokine drugs and other treatment (10). A retrospective clinical study analyzed the clinical data of patients with hematological malignancy who received CD22 CAR T cell therapy and reported that during the active phase of CARHLH, HLH-related cytokine levels are persistently elevated, including IFN-γ, IL-1 and IL-6(11). These cytokines are key contributors to disease progression (11). Rejeski (74) proposed that patients with CARHLH be treated with anakinra in combination with high-dose corticosteroids. Refractory patients can be treated with ruxolitinib and emapalumab therapy (74). Porter et al (64) reported that three patients with CD19 CARHLH demonstrated gradual improvement in laboratory manifestations after treatment with anakinra and/or ruxolitinib. Repeat PET-CT scans in two patients revealed complete remission, but relapse or progression of the disease was observed (64). A clinical study analyzed data from 58 patients who received CD22+ CAR T cell therapy: CRS was observed in 50 patients and HLH-like toxicity was identified in 19 patients (38%). Notably, the toxic manifestations improved in all patients following administration of anakinra or the addition of corticosteroids (13). In one patient, all abnormal laboratory parameters returned to normal following 1 month of treatment with anakinra (13).
SITC guidelines suggest that tocilizumab treatment should be promptly initiated in patients with suspectedCARHLH. Anakinra and corticosteroids may be considered for tocilizumab-refractory HLH/MAS-like toxicity (69). Corticosteroids have side effects, including hypertension, metabolic derangement, fractures and increased risk of infection (75). According to the ASCO guidelines (70), corticosteroids and IL-6 antagonist therapy should be used to manage CARHLH with grade ≥3 organ toxicity. Anakinra is administered to patients with refractory CARHLH (74). Tocilizumab demonstrates limited therapeutic effectiveness in adult patients with sHLH and may elevate the risk of infection-related complications when compared with conventional treatment regimens (76). ASTCT expert recommendations for CARHLH suggest that anakinra and/or corticosteroids are first-line agents (Table V). Anakinra is an IL-1 receptor antagonist; clinical trials of anakinra for sHLH have demonstrated benefit without compromising the efficacy of CAR T cells (77–79). Common toxicities associated with anakinra include infection, headaches and fever. For patients needing further intervention to manage CARHLH, particularly patients with worsening inflammatory markers or signs of progressive end-organ damage, second-line treatments such as ruxolitinib may be considered as a therapeutic option (80). However, ruxolitinib may increase the risk of infection and exacerbate cytopenia and there is still a lack of research and clinical experience (80). Etoposide and emapalumab may serve as alternative treatment options for patients with life-threatening refractory CARHLH (9). Etoposide is the preferred T cell-depleting agent owing to the large amount of data available in both pHLH and sHLH (81–84). The side effects of etoposide include dose-limiting myelosuppression, particularly neutropenia, which may increase the risk of infection (85). Although IFN-γ serves a key role in the development of CARHLH, research on anti-IFN therapy for the treatment of CARHLH is limited (11,86,87). Antagonizing IFN-γ may not affect the efficacy of CAR T cells (86). The application of emapalumab for treating adults with CARHLH is largely speculative. Although emapalumab has been approved by the US Food and Drug Administration for the treatment of pHLH and yields good results, there is debate regarding its efficacy and safety in the treatment of CARHLH (88,89). Currently, targeted therapy is the primary treatment approach for CARHLH. The pathophysiological mechanisms underlying CARHLH are under investigation, and future research may uncover novel mechanisms, facilitating development of targeted therapies based on new molecular targets (19,90). Owing to limited literature on the management of CARHLH and a lack of prospective clinical trials for CARHLH, there is no standardized and unified treatment for CARHLH. However, current guidelines and expert consensus suggest that anti-cytokine therapy serves an important role in the treatment of CARHLH.
Conclusion
CARHLH and CRS exhibit distinct pathogenic mechanisms, although their clinical manifestations are overlapping, with all patients with CARHLH presenting concurrent or prior CRS (8,9,11). The pathogenesis of CARHLH is still being explored. Excessive CAR T cell expansion, NK cell decrease and delayed T cell contraction are the potential mechanisms of action (11,51–53). There are no standardized diagnostic or treatment criteria for CARHLH. ASCO guidelines recommend tocilizumab (an IL-6 monoclonal antibody) for CRS treatment (58). The timing of targeted therapy is determined by the CRS grade (58). During the diagnostic workup for CARHLH, it is key to differentiate whether sHLH is driven by CAR T therapy (59). Regular monitoring of CAR T cell expansion dynamics can aid this distinction (59). Patients at a high risk of HLH can be identified early using indicators such as ferritin and HLH-associated cytokine levels (9). Patients with suspected CARHLH should receive CARHLH treatment as soon as possible (9). Currently, there is no unified therapeutic regimen for CARHLH. The 2023 ASTCT expert consensus recommends anakinra and corticosteroids as first-line agents, while ruxolitinib, etoposide and emapalumab may serve as alternative therapies for refractory cases (58). Corticosteroids, anti-IL-1 and anti-IL-6 therapies are the most frequently used, followed by etoposide (12,50,59,67,68,91–93). Given the abnormal CAR T cell proliferation during CARHLH progression, T cell-targeted therapy could be considered in rapidly progressing or life-threatening scenarios when first- and second-line treatments fail. However, research and clinical experience remain scarce, necessitating further investigation.
In conclusion, the specific pathogenesis of CARHLH is unclear, and the diagnostic and treatment criteria need to be improved and standardized. CARHLH is a rare disease, and the mechanisms of its occurrence and development are not fully understood. Larger multicenter studies need to be conducted or insights should be extrapolated from other sHLH cases to develop diagnosis, treatment and prediction models and systematic understanding of CARHLH.
Acknowledgements
Not applicable.
Funding
The present study was supported by the National Natural Science Foundation of China (grant no. 82170122).
Availability of data and materials
Not applicable.
Authors' contributions
JH, CF, LH and YW wrote the manuscript. CF, LH and YW revised the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Shimabukuro-Vornhagen A, Böll B, Schellongowski P, Valade S, Metaxa V, Azoulay E and von Bergwelt-Baildon M: Critical care management of chimeric antigen receptor T-cell therapy recipients. CA Cancer J Clin. 72:78–93. 2022. View Article : Google Scholar : PubMed/NCBI | |
Lin H, Cheng J, Mu W, Zhou J and Zhu L: Advances in universal CAR-T cell therapy. Front Immunol. 12:7448232021. View Article : Google Scholar : PubMed/NCBI | |
Cameron BJ, Gerry AB, Dukes J, Harper JV, Kannan V, Bianchi FC, Grand F, Brewer JE, Gupta M, Plesa G, et al: Identification of a Titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Sci Transl Med. 5:197ra1032013. View Article : Google Scholar : PubMed/NCBI | |
Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM and Rosenberg SA: 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. View Article : Google Scholar : PubMed/NCBI | |
Brudno JN and Kochenderfer JN: Toxicities of chimeric antigen receptor T cells: Recognition and management. Blood. 127:3321–3330. 2016. View Article : Google Scholar : PubMed/NCBI | |
Owusu KA, Schiffer M and Perreault S: Chimeric antigen receptor T cells: Toxicity and management considerations. AACN Adv Crit Care. 33:301–307. 2022. View Article : Google Scholar : PubMed/NCBI | |
Freyer CW and Porter DL: Cytokine release syndrome and neurotoxicity following CAR T-cell therapy for hematologic malignancies. J Allergy Clin Immunol. 146:940–948. 2020. View Article : Google Scholar : PubMed/NCBI | |
Major A, Collins J, Craney C, Heitman AK, Bauer E, Zerante E, Stock W, Bishop MR and Jasielec J: Management of hemophagocytic lymphohistiocytosis (HLH) associated with chimeric antigen receptor T-cell (CAR-T) therapy using anti-cytokine therapy: An illustrative case and review of the literature. Leuk Lymphoma. 62:1765–1769. 2021. View Article : Google Scholar : PubMed/NCBI | |
Hines MR, Knight TE, McNerney KO, Leick MB, Jain T, Ahmed S, Frigault MJ, Hill JA, Jain MD, Johnson WT, et al: Immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome. Transplant Cell Ther. 29:438.e1–438.e16. 2023. View Article : Google Scholar : PubMed/NCBI | |
Sandler RD, Tattersall RS, Schoemans H, Greco R, Badoglio M, Labopin M, Alexander T, Kirgizov K, Rovira M, Saif M, et al: Diagnosis and management of secondary HLH/MAS following HSCT and CAR-T cell therapy in adults; A review of the literature and a survey of practice within EBMT centres on behalf of the autoimmune diseases working party (ADWP) and transplant complications working party (TCWP). Front Immunol. 11:5242020. View Article : Google Scholar : PubMed/NCBI | |
Lichtenstein DA, Schischlik F, Shao L, Steinberg SM, Yates B, Wang HW, Wang Y, Inglefield J, Dulau-Florea A, Ceppi F, et al: Characterization of HLH-like manifestations as a CRS variant in patients receiving CD22 CAR T cells. Blood. 138:2469–2484. 2021. View Article : Google Scholar : PubMed/NCBI | |
Hines MR, Keenan C, Maron Alfaro G, Cheng C, Zhou Y, Sharma A, Hurley C, Nichols KE, Gottschalk S, Triplett BM and Talleur AC: Hemophagocytic lymphohistiocytosis-like toxicity (carHLH) after CD19-specific CAR T-cell therapy. Br J Haematol. 194:701–707. 2021. View Article : Google Scholar : PubMed/NCBI | |
Shah NN, Highfill SL, Shalabi H, Yates B, Jin J, Wolters PL, Ombrello A, Steinberg SM, Martin S, Delbrook C, et al: CD4/CD8 T-cell selection affects chimeric antigen receptor (CAR) T-cell potency and toxicity: updated results from a phase I anti-CD22 CAR T-cell trial. J Clin Oncol. 38:1938–1950. 2020. View Article : Google Scholar : PubMed/NCBI | |
Debaugnies F, Mahadeb B, Ferster A, Meuleman N, Rozen L, Demulder A and Corazza F: Performances of the H-score for diagnosis of hemophagocytic lymphohistiocytosis in adult and pediatric patients. Am J Clin Pathol. 145:862–870. 2016. View Article : Google Scholar : PubMed/NCBI | |
Schubert ML, Schmitt M, Wang L, Ramos CA, Jordan K, Müller-Tidow C and Dreger P: Side-effect management of chimeric antigen receptor (CAR) T-cell therapy. Ann Oncol. 32:34–48. 2021. View Article : Google Scholar : PubMed/NCBI | |
Hayden PJ, Roddie C, Bader P, Basak GW, Bonig H, Bonini C, Chabannon C, Ciceri F, Corbacioglu S, Ellard R, et al: Management of adults and children receiving CAR T-cell therapy: 2021 Best practice recommendations of the European society for blood and marrow transplantation (EBMT) and the joint accreditation committee of ISCT and EBMT (JACIE) and the European haematology association (EHA). Ann Oncol. 33:259–275. 2022. View Article : Google Scholar : PubMed/NCBI | |
Ponnatt TS, Lilley CM and Mirza KM: Hemophagocytic lymphohistiocytosis. Arch Pathol Lab Med. 146:507–519. 2022. View Article : Google Scholar : PubMed/NCBI | |
Schram AM, Comstock P, Campo M, Gorovets D, Mullally A, Bodio K, Arnason J and Berliner N: Haemophagocytic lymphohistiocytosis in adults: A multicentre case series over 7 years. Br J Haematol. 172:412–419. 2016. View Article : Google Scholar : PubMed/NCBI | |
Henter JI: Hemophagocytic lymphohistiocytosis. N Engl J Med. 392:584–598. 2025. View Article : Google Scholar : PubMed/NCBI | |
Henter JI, Elinder G, Söder O and Ost A: Incidence in Sweden and clinical features of familial hemophagocytic lymphohistiocytosis. Acta Paediatr Scand. 80:428–435. 1991. View Article : Google Scholar : PubMed/NCBI | |
Ravelli A, Minoia F, Davi S, Horne A, Bovis F, Pistorio A, Aricò M, Avcin T, Behrens EM, De Benedetti F, et al: 2016 Classification criteria for macrophage activation syndrome complicating systemic juvenile idiopathic arthritis: A European league against rheumatism/American college of rheumatology/paediatric rheumatology international trials organisation collaborative initiative. Ann Rheum Dis. 75:481–489. 2016. View Article : Google Scholar : PubMed/NCBI | |
Ramos-Casals M, Brito-Zerón P, López-Guillermo A, Khamashta MA and Bosch X: Adult haemophagocytic syndrome. Lancet. 383:1503–1516. 2014. View Article : Google Scholar : PubMed/NCBI | |
He L, Yang C and Wang Y: Biological therapies for hemophagocytic lymphohistiocytosis: Current knowledge and future perspectives. Expert Opin Biol Ther. 23:1005–1013. 2023. View Article : Google Scholar : PubMed/NCBI | |
Janka GE and Lehmberg K: Hemophagocytic syndromes-an update. Blood Rev. 28:135–142. 2014. View Article : Google Scholar : PubMed/NCBI | |
Griffin G, Shenoi S and Hughes GC: Hemophagocytic lymphohistiocytosis: An update on pathogenesis, diagnosis, and therapy. Best Pract Res Clin Rheumatol. 34:1015152020. View Article : Google Scholar : PubMed/NCBI | |
Filipovich A, McClain K and Grom A: Histiocytic disorders: Recent insights into pathophysiology and practical guidelines. Biol Blood Marrow Transplant. 16 (1 Suppl):S82–S89. 2010. View Article : Google Scholar : PubMed/NCBI | |
Arceci RJ: When T cells and macrophages do not talk: The hemophagocytic syndromes. Curr Opin Hematol. 15:359–367. 2008. View Article : Google Scholar : PubMed/NCBI | |
Niece JA, Rogers ZR, Ahmad N, Langevin AM and McClain KL: Hemophagocytic lymphohistiocytosis in Texas: Observations on ethnicity and race. Pediatr Blood Cancer. 54:424–428. 2010. View Article : Google Scholar : PubMed/NCBI | |
Löfstedt A, Jädersten M, Meeths M and Henter JI: Malignancy-associated hemophagocytic lymphohistiocytosis in Sweden: Incidence, clinical characteristics, and survival. Blood. 143:233–242. 2024. View Article : Google Scholar : PubMed/NCBI | |
Singh S, Khasbage S, Kaur RJ, Sidhu JK and Bhandari B: Chimeric antigen receptor T cell: A cancer immunotherapy. Indian J Pharmacol. 54:226–233. 2022. View Article : Google Scholar : PubMed/NCBI | |
Chong EA, Ruella M and Schuster SJ; Lymphoma Program Investigators at the University of Pennsylvania, : Five-year outcomes for refractory B-cell lymphomas with CAR T-cell therapy. N Engl J Med. 384:673–674. 2021. View Article : Google Scholar : PubMed/NCBI | |
Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, Chew A, Gonzalez VE, Zheng Z, Lacey SF, et al: Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 371:1507–1517. 2014. View Article : Google Scholar : PubMed/NCBI | |
Cappell KM, Sherry RM, Yang JC, Goff SL, Vanasse DA, McIntyre L, Rosenberg SA and Kochenderfer JN: Long-term follow-up of anti-CD19 chimeric antigen receptor T-cell therapy. J Clin Oncol. 38:3805–3815. 2020. View Article : Google Scholar : PubMed/NCBI | |
Feins S, Kong W, Williams EF, Milone MC and Fraietta JA: An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am J Hematol. 94((S1)): S3–S9. 2019.PubMed/NCBI | |
Stone JD and Kranz DM: Role of T cell receptor affinity in the efficacy and specificity of adoptive T cell therapies. Front Immunol. 4:2442013. View Article : Google Scholar : PubMed/NCBI | |
Berdeja JG, Madduri D, Usmani SZ, Jakubowiak A, Agha M, Cohen AD, Stewart AK, Hari P, Htut M, Lesokhin A, et al: Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): A phase 1b/2 open-label study: A phase 1b/2 open-label study. Lancet. 398:314–324. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang T, Tang Y, Cai J, Wan X, Hu S, Lu X, Xie Z, Qiao X, Jiang H, Shao J, et al: Coadministration of CD19- and CD22-directed chimeric antigen receptor T-cell therapy in childhood B-cell acute lymphoblastic leukemia: A single-arm, multicenter, phase II trial. J Clin Oncol. 41:1670–1683. 2023. View Article : Google Scholar : PubMed/NCBI | |
Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, Braunschweig I, Oluwole OO, Siddiqi T, Lin Y, et al: Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 377:2531–2544. 2017. View Article : Google Scholar : PubMed/NCBI | |
Schuster SJ, Bishop MR, Tam CS, Waller EK, Borchmann P, McGuirk JP, Jäger U, Jaglowski S, Andreadis C, Westin JR, et al: Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med. 380:45–56. 2019. View Article : Google Scholar : PubMed/NCBI | |
Frey NV, Shaw PA, Hexner EO, Pequignot E, Gill S, Luger SM, Mangan JK, Loren AW, Perl AE, Maude SL, et al: Optimizing chimeric antigen receptor T-cell therapy for adults with acute lymphoblastic leukemia. J Clin Oncol. 38:415–422. 2020. View Article : Google Scholar : PubMed/NCBI | |
Raje N, Berdeja J, Lin Y, Siegel D, Jagannath S, Madduri D, Liedtke M, Rosenblatt J, Maus MV, Turka A, et al: Anti-BCMA CAR T-cell therapy bb2121 in relapsed or refractory multiple myeloma. N Engl J Med. 380:1726–1737. 2019. View Article : Google Scholar : PubMed/NCBI | |
Bupha-Intr O, Haeusler G, Chee L, Thursky K, Slavin M and Teh B: CAR-T cell therapy and infection: A review. Expert Rev Anti Infect Ther. 19:749–758. 2021. View Article : Google Scholar : PubMed/NCBI | |
Lei W, Xie M, Jiang Q, Xu N, Li P, Liang A, Young KH and Qian W: Treatment-related adverse events of chimeric antigen receptor T-cell (CAR T) in clinical trials: A systematic review and meta-analysis. Cancers (Basel). 13:39122021. View Article : Google Scholar : PubMed/NCBI | |
Zu C, Wu S, Zhang M, Wei G, Xu H, Cui J, Chang AH, Huang H and Hu Y: A distinct cytokine network distinguishes chimeric antigen receptor T cell (CAR-T)-associated hemophagocytic lymphohistiocytosis-like toxicity (carHLH) from severe cytokine release syndrome following CAR-T therapy. Cytotherapy. 25:1167–1175. 2023. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Fang Y, Chen X, Wang Z, Liang X, Zhang T, Liu M, Zhou N, Lv J, Tang K, et al: Gasdermin E-mediated target cell pyroptosis by CAR T cells triggers cytokine release syndrome. Sci Immunol. 5:eaax79692020. View Article : Google Scholar : PubMed/NCBI | |
Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, Bader P, Verneris MR, Stefanski HE, Myers GD, et al: Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 378:439–448. 2018. View Article : Google Scholar : PubMed/NCBI | |
Brudno JN and Kochenderfer JN: Current understanding and management of CAR T cell-associated toxicities. Nat Rev Clin Oncol. 21:501–521. 2024. View Article : Google Scholar : PubMed/NCBI | |
Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, Komanduri KV, Lin Y, Jain N, Daver N, et al: Chimeric antigen receptor T-cell therapy-assessment and management of toxicities. Nat Rev Clin Oncol. 15:47–62. 2018. View Article : Google Scholar : PubMed/NCBI | |
Fajgenbaum DC and June CH: Cytokine storm. N Engl J Med. 383:2255–2273. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yan W, Xiong Y, Lv R, Du C, Yu T, Zhang S, Sui W, Deng S, Xiao J, Xu Y, et al: Uncommon biphasic CAR-T expansion induces hemophagocytic lymphohistiocytosis-like syndrome and fatal multiple infections following BCMA CAR-T cell therapy: A case report. J Immunother Cancer. 12:e0100802024. View Article : Google Scholar : PubMed/NCBI | |
Vandenhaute J, Wouters CH and Matthys P: Natural killer cells in systemic autoinflammatory diseases: A focus on systemic juvenile idiopathic arthritis and macrophage activation syndrome. Front Immunol. 10:30892020. View Article : Google Scholar : PubMed/NCBI | |
Weiss ES, Girard-Guyonvarc'h C, Holzinger D, de Jesus AA, Tariq Z, Picarsic J, Schiffrin EJ, Foell D, Grom AA, Ammann S, et al: Interleukin-18 diagnostically distinguishes and pathogenically promotes human and murine macrophage activation syndrome. Blood. 131:1442–1455. 2018. View Article : Google Scholar : PubMed/NCBI | |
Kaplanski G: Interleukin-18: Biological properties and role in disease pathogenesis. Immunol Rev. 281:138–153. 2018. View Article : Google Scholar : PubMed/NCBI | |
Miao L, Zhang Z, Ren Z and Li Y: Reactions related to CAR-T cell therapy. Front Immunol. 12:6632012021. View Article : Google Scholar : PubMed/NCBI | |
Nichols KE and Hines MR: NK cells: Energized yet exhausted in adult HLH. Blood. 136:524–525. 2020. View Article : Google Scholar : PubMed/NCBI | |
Strati P, Ahmed S, Kebriaei P, Nastoupil LJ, Claussen CM, Watson G, Horowitz SB, Brown ART, Do B, Rodriguez MA, et al: Clinical efficacy of anakinra to mitigate CAR T-cell therapy-associated toxicity in large B-cell lymphoma. Blood Adv. 4:3123–3127. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ahmed S, Furqan F, Strati P, Westin J, Fayad LE, Hagemeister FB, Lee HJ, Iyer SP, Nair R, Nastoupil LJ, et al: Haemophagocytic lymphohistiocytosis (HLH) in patients with large B-cell lymphoma treated with standard of care (SOC) axicabtagene ciloleucel (Axi-cel). J Clin Oncol. 38 (15 Suppl):S80572020. View Article : Google Scholar | |
Lee DW, Santomasso BD, Locke FL, Ghobadi A, Turtle CJ, Brudno JN, Maus MV, Park JH, Mead E, Pavletic S, et al: ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant. 25:625–638. 2019. View Article : Google Scholar : PubMed/NCBI | |
Meireles AM, Iacoboni G, Moço LM, Ramos I, Brás G, Azevedo J, Rodrigues Â, Moreira C and Mariz M: Hemophagocytic lymphohistiocytosis in a patient with Epstein-Barr virus-positive diffuse large B-cell lymphoma treated with chimeric antigen receptor T-cell therapy. Immunotherapy. 16:1105–1111. 2024. View Article : Google Scholar : PubMed/NCBI | |
Khurana A, Rosenthal AC, Mohty R, Gaddam M, Bansal R, Hathcock MA, Nedved AN, Durani U, Iqbal M, Wang Y, et al: Chimeric antigen receptor T-cell therapy associated hemophagocytic lymphohistiocytosis syndrome: Clinical presentation, outcomes, and management. Blood Cancer J. 14:1362024. View Article : Google Scholar : PubMed/NCBI | |
Henter JI, Horne A, Aricó M, Egeler RM, Filipovich AH, Imashuku S, Ladisch S, McClain K, Webb D, Winiarski J and Janka G: HLH-2004: Diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 48:124–131. 2007. View Article : Google Scholar : PubMed/NCBI | |
Fardet L, Galicier L, Lambotte O, Marzac C, Aumont C, Chahwan D, Coppo P and Hejblum G: Development and validation of the HScore, a score for the diagnosis of reactive hemophagocytic syndrome. Arthritis Rheumatol. 66:2613–2620. 2014. View Article : Google Scholar : PubMed/NCBI | |
Takagi S, Masuoka K, Uchida N, Ishiwata K, Araoka H, Tsuji M, Yamamoto H, Kato D, Matsuhashi Y, Kusumi E, et al: High incidence of haemophagocytic syndrome following umbilical cord blood transplantation for adults. Br J Haematol. 147:543–553. 2009. View Article : Google Scholar : PubMed/NCBI | |
Porter TJ, Lazarevic A, Ziggas JE, Fuchs E, Kim K, Byrnes H, Luznik L, Bolaños-Meade J, Ali SA, Shah NN, et al: Hyperinflammatory syndrome resembling haemophagocytic lymphohistiocytosis following axicabtagene ciloleucel and brexucabtagene autoleucel. Br J Haematol. 199:720–727. 2022. View Article : Google Scholar : PubMed/NCBI | |
Turtle CJ, Hanafi LA, Berger C, Hudecek M, Pender B, Robinson E, Hawkins R, Chaney C, Cherian S, Chen X, 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. 8:355ra1162016. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Zhou F, Wu Z, Li Y, Li C, Du M, Luo W, Kou H, Lu C and Mei H: Timing of tocilizumab administration under the guidance of IL-6 in CAR-T therapy for R/R acute lymphoblastic leukemia. Front Immunol. 13:9149592022. View Article : Google Scholar : PubMed/NCBI | |
He J, Xu N, Zhou H, Zhou Y, Wu D, Zhao R, Lin T, Xu J, Cao R, Li P and Liu Q: Case report: Chimeric antigen receptor T cells induced late severe cytokine release syndrome. Front Oncol. 12:8939282022. View Article : Google Scholar : PubMed/NCBI | |
Sigha OB, Mbono Betoko R, Nkoro GA, Fossi Happi M, Ekoube CE, Kelbaba BB, Mandeng Ma Linwa E and Kouotou EA: Bart's syndrome associated with a disorder of sexual differentiation: An atypical presentation in a Cameroonian newborn. Clin Case Rep. 10:e052342022. View Article : Google Scholar : PubMed/NCBI | |
Maus MV, Alexander S, Bishop MR, Brudno JN, Callahan C, Davila ML, Diamonte C, Dietrich J, Fitzgerald JC, Frigault MJ, et al: Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune effector cell-related adverse events. J Immunother Cancer. 8:e0015112020. View Article : Google Scholar : PubMed/NCBI | |
Santomasso BD, Nastoupil LJ, Adkins S, Lacchetti C, Schneider BJ, Anadkat M, Atkins MB, Brassil KJ, Caterino JM, Chau I, et al: Management of immune-related adverse events in patients treated with chimeric antigen receptor T-cell therapy: ASCO guideline. J Clin Oncol. 39:3978–3992. 2021. View Article : Google Scholar : PubMed/NCBI | |
Henter JI, Aricò M, Egeler RM, Elinder G, Favara BE, Filipovich AH, Gadner H, Imashuku S, Janka-Schaub G, Komp D, et al: HLH-94: A treatment protocol for hemophagocytic lymphohistiocytosis. HLH study group of the histiocyte society. Med Pediatr Oncol. 28:342–347. 1997. View Article : Google Scholar : PubMed/NCBI | |
Locatelli F, Jordan MB, Allen C, Cesaro S, Rizzari C, Rao A, Degar B, Garrington TP, Sevilla J, Putti MC, et al: Emapalumab in children with primary hemophagocytic lymphohistiocytosis. N Engl J Med. 382:1811–1822. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ge J, Zhang Q, Ma H, Wang D, Zhao Y, Zhu T, Wang W, Zhou C, Wei A, Lian H, et al: Ruxolitinib-based regimen in children with primary hemophagocytic lymphohistiocytosis. Haematologica. 109:458–465. 2024. View Article : Google Scholar : PubMed/NCBI | |
Rejeski K, Subklewe M, Aljurf M, Bachy E, Balduzzi A, Barba P, Bruno B, Benjamin R, Carrabba MG, Chabannon C, et al: Immune effector cell-associated hematotoxicity: EHA/EBMT consensus grading and best practice recommendations. Blood. 142:865–877. 2023. View Article : Google Scholar : PubMed/NCBI | |
Curtis JR, Westfall AO, Allison J, Bijlsma JW, Freeman A, George V, Kovac SH, Spettell CM and Saag KG: Population-based assessment of adverse events associated with long-term glucocorticoid use. Arthritis Rheum. 55:420–426. 2006. View Article : Google Scholar : PubMed/NCBI | |
Kim JY, Kim M, Park JK, Lee EB, Park JW and Hong J: Limited efficacy of tocilizumab in adult patients with secondary hemophagocytic lymphohistiocytosis: A retrospective cohort study. Orphanet J Rare Dis. 17:3632022. View Article : Google Scholar : PubMed/NCBI | |
Park JH, Nath K, Devlin SM, Sauter CS, Palomba ML, Shah G, Dahi P, Lin RJ, Scordo M, Perales MA, et al: CD19 CAR T-cell therapy and prophylactic anakinra in relapsed or refractory lymphoma: Phase 2 trial interim results. Nat Med. 29:1710–1717. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wohlfarth P, Agis H, Gualdoni GA, Weber J, Staudinger T, Schellongowski P and Robak O: Interleukin 1 receptor antagonist anakinra, intravenous immunoglobulin, and corticosteroids in the management of critically ill adult patients with hemophagocytic lymphohistiocytosis. J Intensive Care Med. 34:723–731. 2019. View Article : Google Scholar : PubMed/NCBI | |
Diorio C, Vatsayan A, Talleur AC, Annesley C, Jaroscak JJ, Shalabi H, Ombrello AK, Hudspeth M, Maude SL, Gardner RA and Shah NN: Anakinra utilization in refractory pediatric CAR T-cell associated toxicities. Blood Adv. 6:3398–3403. 2022. View Article : Google Scholar : PubMed/NCBI | |
Abedin S, McKenna E, Chhabra S, Pasquini M, Shah NN, Jerkins J, Baim A, Runaas L, Longo W, Drobyski W, et al: Efficacy, toxicity, and infectious complications in ruxolitinib-treated patients with corticosteroid-refractory graft-versus-host disease after hematopoietic cell transplantation. Biol Blood Marrow Transplant. 25:1689–1694. 2019. View Article : Google Scholar : PubMed/NCBI | |
Bracaglia C, de Graaf K, Pires Marafon D, Guilhot F, Ferlin W, Prencipe G, Caiello I, Davì S, Schulert G, Ravelli A, et al: Elevated circulating levels of interferon-γ and interferon-γ-induced chemokines characterise patients with macrophage activation syndrome complicating systemic juvenile idiopathic arthritis. Ann Rheum Dis. 76:166–172. 2017. View Article : Google Scholar : PubMed/NCBI | |
Horne A, von Bahr Greenwood T, Chiang SCC, Meeths M, Björklund C, Ekelund M, Erensjö P, Berg S, Hagelberg S, Bryceson YT, et al: Efficacy of moderately dosed etoposide in macrophage activation syndrome-hemophagocytic lymphohistiocytosis. J Rheumatol. 48:1596–1602. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zondag TCE, Lika A and van Laar JAM: The role of etoposide in the treatment of adult patients with hemophagocytic lymphohistiocytosis. Exp Hematol Oncol. 12:22023. View Article : Google Scholar : PubMed/NCBI | |
Song Y, Wang J, Wang Y, Wu L and Wang Z: Requirement for containing etoposide in the initial treatment of lymphoma associated hemophagocytic lymphohistiocytosis. Cancer Biol Ther. 22:598–606. 2021. View Article : Google Scholar : PubMed/NCBI | |
Henter JI, von Bahr Greenwood T and Bergsten E: Emapalumab in primary hemophagocytic lymphohistiocytosis. N Engl J Med. 383:596–598. 2020. View Article : Google Scholar : PubMed/NCBI | |
Bailey SR, Vatsa S, Larson RC, Bouffard AA, Scarfò I, Kann MC, Berger TR, Leick MB, Wehrli M, Schmidts A, et al: Blockade or deletion of IFNγ reduces macrophage activation without compromising CAR T-cell function in hematologic malignancies. Blood Cancer Discov. 3:136–153. 2022. View Article : Google Scholar : PubMed/NCBI | |
McNerney KO, DiNofia AM, Teachey DT, Grupp SA and Maude SL: Potential role of IFNγ inhibition in refractory cytokine release syndrome associated with CAR T-cell therapy. Blood Cancer Discov. 3:90–94. 2022. View Article : Google Scholar : PubMed/NCBI | |
Verkamp B, Jodele S, Sabulski A, Marsh RA, Kieser P and Jordan MB: Emapalumab therapy for hemophagocytic lymphohistiocytosis before reduced-intensity transplantation improves chimerism. Blood. 144:2625–2636. 2024. View Article : Google Scholar : PubMed/NCBI | |
Garonzi C, Chinello M and Cesaro S: Emapalumab for adult and pediatric patients with hemophagocytic lymphohistiocytosis. Expert Rev Clin Pharmacol. 14:527–534. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wu Y, Sun X, Kang K, Yang Y, Li H, Zhao A and Niu T: Hemophagocytic lymphohistiocytosis: Current treatment advances, emerging targeted therapy and underlying mechanisms. J Hematol Oncol. 17:1062024. View Article : Google Scholar : PubMed/NCBI | |
Roddie C, Lekakis LJ, Marzolini MAV, Ramakrishnan A, Zhang Y, Hu Y, Peddareddigari VGR, Khokhar N, Chen R, Basilico S, et al: Dual targeting of CD19 and CD22 with bicistronic CAR-T cells in patients with relapsed/refractory large B-cell lymphoma. Blood. 141:2470–2482. 2023.PubMed/NCBI | |
Schultz LM, Jeyakumar N, Kramer AM, Sahaf B, Srinagesh H, Shiraz P, Agarwal N, Hamilton M, Erickson C, Jacobs A, et al: CD22 CAR T cells demonstrate high response rates and safety in pediatric and adult B-ALL: Phase 1b results. Leukemia. 38:963–968. 2024. View Article : Google Scholar : PubMed/NCBI | |
Masih KE, Ligon JA, Yates B, Shalabi H, Little L, Islam Z, Ombrello AK, Inglefield J, Nussenblatt V, Manion M, et al: Consequences of hemophagocytic lymphohistiocytosis-like cytokine release syndrome toxicities and concurrent bacteremia. Pediatr Blood Cancer. 68:e292472021. View Article : Google Scholar : PubMed/NCBI | |
Service USDoHaH, Health NIo and Institute NC: National Cancer Institute, . Common terminology criteria for adverse events (CTCAE). Version 5.0.2017. |