Open Access

Efficacy of emapalumab in the management of anti‑CD19 chimeric antigen receptor T‑cell therapy‑associated cytokine release syndrome: A report of two cases

  • Authors:
    • Wenzhi Cai
    • Yutong Lu
    • Haiju He
    • Jiaqi Li
    • Shuangzhu Liu
    • Hongzhi Geng
    • Qin Yang
    • Liangyu Zeng
    • Depei Wu
    • Caixia Li
  • View Affiliations

  • Published online on: November 22, 2024     https://doi.org/10.3892/ol.2024.14817
  • Article Number: 71
  • Copyright: © Cai et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Chimeric antigen receptor (CAR) T‑cell therapy is an effective treatment for diffuse large B‑cell lymphoma (DLBCL). However, it may activate the systemic immune system of the patient, resulting in cytokine release syndrome (CRS). Emapalumab is a human monoclonal antibody targeting interferon‑γ, inhibiting its interaction with cell surface receptors and the subsequent activation of inflammatory pathways. The present report describes the cases of 2 patients with relapsed DLBCL treated with CAR T‑cell therapy, in which the severe CRS associated with CAR T‑cell therapy was attenuated without compromising antitumor efficacy after receiving emapalumab. Further prospective clinical trials are warranted to determine the role of emapalumab in CAR T‑cell therapy.

Introduction

In total, 20–50% of patients with diffuse large B-cell lymphoma (DLBCL) will become resistant to initial immuno-chemotherapy or relapse after achieving a complete metabolic response (CMR), thus predicting a poor prognosis (1). Chimeric antigen receptor (CAR) T-cell therapy targeting CD19 has resulted in tremendous progress with regard to increasing survival rates in the past two decades (25). In the ZUMA-1 trial, patients with refractory large B-cell lymphoma received axicabtagene ciloleucel autologous anti-CD19 CAR T-cell therapy and the long-term survival analysis yielded an estimated 5-year OS rate of 42.6% (5), suggesting that further investigations are warranted to maintain long-term clinical remission, as well as to manage the potential infusion-related toxicities, namely cytokine release syndrome (CRS; grade ≥3 reported in 2–22% of patients) and immune effector cell-associated neurotoxicity syndrome (grade ≥3 reported in 5.1–28% of patients).

Cytokine release syndrome (CRS) is a potentially life threatening toxicity with a prevalence that can be as high as 93% (6,7). Currently, the commonly used treatment regimens for CRS include tocilizumab and corticosteroids (8). In patients with CRS who respond to tocilizumab, symptoms such as fever and hypotension typically resolve within a few hours. However, a subset of patients may show no improvement in symptoms or response. In these cases, the consideration of other drugs, such as corticosteroids, is warranted (9,10). Nevertheless, some studies have indicate that corticosteroids may adversely affect CAR T-cell function, and the cumulative dose could be linked to reduced survival rates (9,11,12). Additionally, some patients exhibit poor responses to both treatment regimens, resulting in limited options. Consequently, there is an urgent need to explore other medications for more effective management (10,13). The present study presented two cases of patients with relapsed DLBCL who received CAR T-cell therapy and aimed to investigate the efficacy of the interferon-γ (IFN-γ) antibody emapalumab in treating CAR T-cell induced CRS.

Case report

Patient 1

A 56-year-old male patient was diagnosed with germinal center B-cell (GCB) like DLBCL, Ann Arbor stage IV, an International Prognostic Index score (14) of 3 and MYC-BCL2 double expression in January 2022 at the Department of Oncology, The First Affiliated Hospital of Soochow University. The patient received 8 cycles (every 21 days for a cycle) of R-CHOP (iv rituximab, 375 mg/m2 on day 0; iv cyclophosphamide, 750 mg/m2 on day 1; iv doxorubicin, 50 mg/m2 on day 1; iv vincristine, 1.4 mg/m2, dose cap of 2 mg on day 1; and oral prednisone, 100 mg daily on days 1–5). The fluorodeoxyglucose-positron emission tomography (FDG-PET) scan showed progression in both lungs, with a Deauville score (DS) (15) of 5 and an abdominal node with a DS of 4. A salvage chemotherapy comprising 2 cycles (every 21 days for a cycle) of R-GDP (iv rituximab, 375 mg/m2 on day 0; iv gemcitabine, 1,000 mg/m2 on days 1 and 8; iv dexamethasone, 40 mg on days 1–4; and iv cisplatin, 25 mg/m2 on days 1–3) was administered. The patient was later referred to the National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University for CD19-directed CAR T-cell therapy (16,17) in October 2022. The patient underwent lymphodepleting conditioning with cyclophosphamide 0.3 g/m2 day-5-2 and fludarabine 30 mg/m2 d-5-2. The patient was infused with 2×106 axicabtagene ciloleucel cells/kg. On day 4, the patient experienced a fever of 38.0°C with tachycardia, but was normotensive with a normal respiratory exam. The patient had repeated negative blood cultures and was diagnosed with CRS (grade 1) based on the American Society for Transplantation and Cellular Therapy grading scale (18). Blood cultures testing for pathogenic microorganisms were obtained which was later reported to be negative, and broad-spectrum antibiotics (iv meropenem 1g q8h, and oral voriconazole 4 mg/kg) were administered. On day 6, the patient developed grade 3 CRS with hypotension (85/54 mmHg) and hypoxia. Supplemental oxygen was supplied at 6 l/min, as well as norepinephrine infusions initially at 0.15 µg/kg/min. Two doses of 8 mg/kg tocilizumab were administered intravenously every 12 h, but the patient did not exhibit a notable and persistent change in the fever or blood pressure. The temperature climbed back up to 39.6°C and the blood pressure was 101/62 mmHg treated with norepinephrine infusions. Considering the lack of improvement in CRS symptoms, additional treatment is needed. Due to an increase in the level of IFN-γ, emapalumab, an anti-IFN-γ antibody, was administered at a dose of 1 mg/kg on day 7 after informed consent was obtained. The temperature of the patient rapidly normalized and the norepinephrine dose was gradually reduced (Fig. 1A). The levels of C-reactive protein (CRP) and inflammatory cytokines significantly decreased, with IFN-γ level particularly decreased to 0 pg/ml (Fig. 1B and C). The level of interleukin 6 (IL-6) descended from 1,162 on day 7 to 304 pg/ml on day 8 and continued to fall (Fig. 1C). The expansion of CAR-T cells were monitored by flow cytometry (Fig. 1D): The PE-labeled monoclonal anti-FMC63 antibody (ACROBiosystems, FM3-HPY53), CD3-FITC-conjugated antibody (Immunotech S.A.S, A07746), and CD45-PC7-conjugated antibody (Immunotech S.A.S, IM3548) were used to label CAR-T cells. The Beckman Coulter Navios (Beckman Coulter, Inc.) was used to acquire the data, and Kaluza Analysis Software (v2.1; Beckman Coulter, Inc.) was employed for data analysis. The number of CAR-T cells reached a peak less than 10 days after cell infusion (Fig. 1E). This also suggested that neutralizing IFN-γ may not affect the normal activity of CAR-T cells (19). The patient was followed up at 1, 3, and 6 months after treatment. At 3 months, the patient underwent an FDG-PET scan, showing a partial response (PR) status (16) (Fig. 1F). The response status remained stable at the last follow-up (at 6 months).

Patient 2

A 49-year-old female patient was diagnosed with DLBCL transformed from follicular lymphoma (FL; grade 3B, WHO grading system for FL) (20) in November 2020 at the First People's Hospital of Changzhou. The patient achieved a complete metabolic response (CMR) after 4 cycles of R-CHOP (as above) and 4 cycles of R-CHOP (as above) with zanubrutinib (oral, 160 mg, bid), but relapsed 6 months later. She was referred to the National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, in January 2023. The refractory disease was confirmed with a biopsy of the inguinal lymph node. Next-generation sequencing (NGS) showed an MCD subtype (a genetic subtype in DLBCL, based on the co-occurrence of MYD88L265P and CD79B mutations) (21), with positivity for protein coding gene CD79B, CDKN2A (multiple tumor suppressor l), CCND3 (protein coding gene), FAS (protein coding gene), APC (tumor suppressor gene), B-cell lymphoma 6 (BCL6) and CREBBP (protein coding gene). NGS was performed using a NovaSeq 6000 S1 Reagent Kit (cat. no. 20028319; Illumina, Inc.) and a GeneJET Genomic DNA Purification Kit (cat. no. K0721; Thermo Fisher Scientific, Inc.) to prepare the DNA sample. A Bioanalyzer 2100 (Agilent Technologies, Inc.) was used to verify the quality of the processed samples. The purified library was quantified using the KAPA Library Quantification Kit (KAPA Biosystems; cat. no. 07960140001; Roche Diagnostics). Enriched libraries were amplified and subjected to NGS on Illumina novaseq 6000 platforms (150 cycles; cat. no. 2002746; Illumina, Inc.). The quantitative concentration of the final library was 2 nM. The 2×150 bp paired-end sequencing was performed in a testing laboratory (Geneseeq Technology, Inc.) accredited by the Clinical Laboratory Improvement Amendments and the College of American Pathologists. Data analysis was performed using Trimmomatic 0.39 (https://github.com/usadellab/Trimmomatic).

After oral treatment with 3 cycles of R-ICE (iv rituximab, 375 mg/m2 on day 0; iv ifosfamide, 5,000 mg/m2 on day 2; iv etoposide, 100 mg/m2 on days 1–3; carboplatin area under the curve 5, maximum dose of 800 mg, iv on day 2) and zanubrutinib (as above), the patient proceeded to undergo autologous stem cell transplant (ASCT) followed by CAR T-cell therapy. Pre-treatment with CEAC regimen (oral lomustine, 200 mg/m2 on day 6; iv etoposide, 100 mg/m2 q12h on days-5 to −2; iv cytarabine, 200 mg/m2 q12h on days-5 to −2; and iv cyclophosphamide 1.5 g/m2 on days-5 to −2) and iv cladribine (50 mg/m2 on days-5 to −2) was administered, and autologous hematopoietic stem cells were infused (mononuclear cells, 6.8×108/kg; CD34+ cells, 8.0×106/kg). After 7 days, the patient received a total amount of 100×106 relmacabtagene autoleucel cells/kg. A fever (39.6°C) and hypoxemia developed on day 2 after CAR T-cell infusion, with negative repeat blood cultures, and the patient was diagnosed with rapid progression to grade 2 CRS (18). The serum IL-6 level reached a maximum of 506 pg/ml and the IFN-γ level reached 888 pg/ml. One dose of emapalumab (1 mg/kg) was administered after informed consent was obtained, as well as antifebrile therapy and supplemental oxygen. The body temperature rapidly decreased to 38.7°C (Fig. 2A), with IFN-γ and IL-6 levels rapidly decreasing to 0 pg/ml and 141 pg/ml on day 4 after CAR T-cell infusion, respectively (Fig. 2B and C). The CAR T-cells continued to expand (Fig. 2D and E). The patient was followed up at 1, 3, and 6 months after treatment. The 1-month FDG-PET scan showed a CMR (Fig. 2F), and circulating tumor DNA monitoring at 2 months revealed a negative disease state (Fig. 2G). The patient was still alive, taking zanubrutinib 160 mg bid orally, with no signs of occurrence at the 6-month follow-up.

Discussion

The pathophysiology underpinning the development of CRS stems from activation of immune cells of the tumor microenvironment, directly resulting in excessive production of inflammatory cytokines and chemokines (22). The most commonly affected cytokines are IL-6, IFN-γ, tumor necrosis factor-α, CRP and ferritin (23), which in turn lead to endothelial injury and tissue damage. IL-6 serves a crucial role in mediating CRS, and tocilizumab (anti-IL-6 receptor) has been approved for the management of CRS (24). However, some patients exhibit a poor response to tocilizumab and glucocorticoids (9,10), necessitating adjustments to their treatment regimen based on individual responses. Additionally, there is currently no standardized treatment protocol for CRS.

Some patients with severe CRS exhibit characteristics similar to those of hemophagocytic lymphohistiocytosis (HLH) (10). Notably, IFN-γ levels are elevated in both severe CRS and HLH, and in HLH, IFN-γ is regarded as a key driver of inflammation (10). A maximum fold-change in IFN-γ of >100 has been used to predict grade 3+ CRS, with a sensitivity of 83% and a specificity of 100% (25). The knockdown of IFN-γ preserves the benefits of anti-CD19 CAR T-cell therapy and inhibits the release of multiple cytokines from CAR T-cells and peripheral blood mononuclear cells, thereby reducing the effect of CRS and enhances the safety profile of CAR T-cell therapy (26). In addition, IFN-γ blockade can reduce macrophage activation and improve CAR T-cell function while reducing treatment-related toxicity in hematologic malignancies (27).

Emapalumab is a human anti-IFN-γ antibody that binds to both free and receptor-bound IFN-γ, inhibiting receptor dimerization and the transduction of IFN-γ signaling, thereby neutralizing its biological activity (28). A prospective clinical study has demonstrated its effectiveness as a targeted therapy for primary HLH (28). Additionally, it has shown efficacy in treating CRS following CAR T-cell therapy in patients with relapsed B-cell acute lymphoblastic leukemia (29). McNerney and DiNofia reported the case of a patient with B-cell acute lymphoblastic leukemia who developed grade 4 CRS that was refractory to both tocilizumab and glucocorticoids. This patient was treated with tocilizumab, methylprednisolone, siltuximab and the IFN-γ inhibitor emapalumab, achieving complete remission for 12 months (10). Rainone et al (13) described a case of CAR T-cell therapy-associated macrophage activation syndrome/HLH that was successfully treated with emapalumab in combination with anakinra and corticosteroids.

In the present study, patient 1 developed severe CRS and received tocilizumab, but did not respond adequately, ultimately achieving a PR with emapalumab. In case 2, the IFN-γ levels were elevated, up to 888 pg/ml. A study has indicated that elevated IFN-γ levels are associated with severe CRS (30). Therefore, the patient was directly treated with emapalumab and achieved a favorable response. These findings are consistent with the limited previously reported cases (10,13), suggesting that the clinical use of emapalumab can effectively mitigate severe CRS in CAR T-cell-treated patients without compromising antitumor efficacy. Common AEs associated with emapalumab include infection, hypertension and infusion-related reactions (31). However, the 2 patients in the present report had no obvious AEs and the treatment was well tolerated. The patients felt no discomfort or dissatisfaction during the entire treatment. The cost of the medication was acceptable to the patients and they remained proactive and cooperative throughout the treatment. However, the present study also had certain limitations. It only reported two patients and lacked large-scale statistical data, and further prospective clinical trials are necessary to confirm the long-term efficacy and safety of emapalumab.

In summary, the present study trialed the use of the drug emapalumab to neutralize IFN-γ, in order to mitigate CRS while maintaining the efficacy of CAR T-cell proliferation. Prospective clinical trials are warranted to determine its role in CAR T-cell therapy.

Acknowledgements

Not applicable.

Funding

The present study was supported by a grant from the National Natural Science Foundation of China (grant no. 82000158).

Availability of data and materials

The datasets generated and/or analyzed during the current study are not publicly available to protect patient privacy but are available from the corresponding author on reasonable request.

Authors' contributions

CL and DW were responsible for conception and design. WC and YL were responsible for providing study materials or patients. Collection and assembly of data was performed by HG, QY and LZ. Data analysis and interpretation was performed by WC and YL. HH, JL and SL advised on patient treatment, drafted the discussion and confirmed the authenticity of all the raw data. All authors helped to write the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of the First Affiliated Hospital of Soochow University (Suzhou, China; approval number 2017-053-2).

Patient consent for publication

Written informed consent was obtained from the patients for the publication of any potentially identifiable images or data included in this article.

Competing interests

The authors declare that they have no competing interests.

References

1 

Crump M, Neelapu SS, Farooq U, Van Den Neste E, Kuruvilla J, Westin J, Link BK, Hay A, Cerhan JR, Zhu L, et al: Outcomes in refractory diffuse large B-cell lymphoma: Results from the international SCHOLAR-1 study. Blood. 130:1800–1808. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Locke FL, Ghobadi A, Jacobson CA, Miklos DB, Lekakis LJ, Oluwole OO, Lin Y, Braunschweig I, Hill BT, Timmerman JM, et al: Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): A single-arm, multicentre, phase 1–2 trial. Lancet Oncol. 20:31–42. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Abramson JS, Palomba ML, Gordon LI, Lunning MA, Wang M, Arnason J, Mehta A, Purev E, Maloney DG, Andreadis C, et al: Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): A multicentre seamless design study. Lancet. 396:839–852. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Ying Z, Yang H, Guo Y, Li W, Zou D, Zhou D, Wang Z, Zhang M, Wu J, Liu H, et al: Long-term outcomes of relmacabtagene autoleucel in Chinese patients with relapsed/refractory large B-cell lymphoma: Updated results of the RELIANCE study. Cytotherapy. 25:521–529. 2023. View Article : Google Scholar : PubMed/NCBI

5 

Neelapu SS, Jacobson CA, Ghobadi A, Miklos DB, Lekakis LJ, Oluwole OO, Lin Y, Braunschweig I, Hill BT, Timmerman JM, et al: Five-year follow-up of ZUMA-1 supports the curative potential of axicabtagene ciloleucel in refractory large B-cell lymphoma. Blood. 141:2307–2315. 2023.PubMed/NCBI

6 

Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, Grupp SA and Mackall CL: Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 124:188–195. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Topp MS, van Meerten T, Houot R, Minnema MC, Bouabdallah K, Lugtenburg PJ, Thieblemont C, Wermke M, Song KW, Avivi I, et al: Earlier corticosteroid use for adverse event management in patients receiving axicabtagene ciloleucel for large B-cell lymphoma. Br J Haematol. 195:388–398. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Li P, Liu Y, Liang Y, Bo J, Gao S, Hu Y, Hu Y, Huang H, Huang X, Jing H, et al: 2022 Chinese expert consensus and guidelines on clinical management of toxicity in anti-CD19 chimeric antigen receptor T-cell therapy for B-cell non-Hodgkin lymphoma. Cancer Biol Med. 20:129–146. 2023. View Article : Google Scholar : PubMed/NCBI

9 

Hughes AD, Teachey DT and Diorio C: Riding the storm: Managing cytokine-related toxicities in CAR-T cell therapy. Semin Immunopathol. 46:52024. View Article : Google Scholar : PubMed/NCBI

10 

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

11 

Strati P, Ahmed S, Furqan F, Fayad LE, Lee HJ, Iyer SP, Nair R, Nastoupil LJ, Parmar S, Rodriguez MA, et al: Prognostic impact of corticosteroids on efficacy of chimeric antigen receptor T-cell therapy in large B-cell lymphoma. Blood. 137:3272–3276. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, Chung SS, Stefanski J, Borquez-Ojeda O, Olszewska M, et al: Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 6:224ra2252014. View Article : Google Scholar : PubMed/NCBI

13 

Rainone M, Ngo D, Baird JH, Budde LE, Htut M, Aldoss I and Pullarkat V: Interferon-γ blockade in CAR T-cell therapy-associated macrophage activation syndrome/hemophagocytic lymphohistiocytosis. Blood Adv. 7:533–536. 2023. View Article : Google Scholar : PubMed/NCBI

14 

Sehn LH and Salles G: Diffuse large B-cell lymphoma. N Engl J Med. 384:842–858. 2021. View Article : Google Scholar : PubMed/NCBI

15 

Meignan M, Gallamini A, Meignan M, Gallamini A and Haioun C: Report on the first international workshop on interim-PET-scan in lymphoma. Leuk Lymphoma. 50:1257–1260. 2009. View Article : Google Scholar : PubMed/NCBI

16 

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

17 

Ying Z, Yang H, Guo Y, Li W, Zou D, Zhou D, Wang Z, Zhang M, Wu J, Liu H, et al: Relmacabtagene autoleucel (relma-cel) CD19 CAR-T therapy for adults with heavily pretreated relapsed/refractory large B-cell lymphoma in China. Cancer Med. 10:999–1011. 2021. View Article : Google Scholar : PubMed/NCBI

18 

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

19 

Manni S, Del Bufalo F, Merli P, Silvestris DA, Guercio M, Caruso S, Reddel S, Iaffaldano L, Pezzella M, Di Cecca S, et al: Neutralizing IFNγ improves safety without compromising efficacy of CAR-T cell therapy in B-cell malignancies. Nat Commun. 14:34232023. View Article : Google Scholar : PubMed/NCBI

20 

Carbone A, Roulland S, Gloghini A, Younes A, von Keudell G, López-Guillermo A and Fitzgibbon J: Follicular lymphoma. Nat Rev Dis Primers. 5:832019. View Article : Google Scholar : PubMed/NCBI

21 

Schmitz R, Wright GW, Huang DW, Johnson CA, Phelan JD, Wang JQ, Roulland S, Kasbekar M, Young RM, Shaffer AL, et al: Genetics and pathogenesis of diffuse large B-cell lymphoma. N Engl J Med. 378:1396–1407. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Norelli M, Camisa B, Barbiera G, Falcone L, Purevdorj A, Genua M, Sanvito F, Ponzoni M, Doglioni C, Cristofori P, et al: Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med. 24:739–748. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Maude SL, Barrett D, Teachey DT and Grupp SA: Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J. 20:119–122. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Kotch C, Barrett D and Teachey DT: Tocilizumab for the treatment of chimeric antigen receptor T cell-induced cytokine release syndrome. Expert Rev Clin Immunol. 15:813–822. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, Fry TJ, Orentas R, Sabatino M, Shah NN, 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. View Article : Google Scholar : PubMed/NCBI

26 

Zhang H, Lv X, Kong Q and Tan Y: IL-6/IFN-γ double knockdown CAR-T cells reduce the release of multiple cytokines from PBMCs in vitro. Hum Vaccin Immunother. 18:1–14. 2022. View Article : Google Scholar

27 

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

28 

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

29 

Schuelke MR, Bassiri H, Behrens EM, Canna S, Croy C, DiNofia A, Gollomp K, Grupp S, Lambert M, Lambrix A, et al: Emapalumab for the treatment of refractory cytokine release syndrome in pediatric patients. Blood Adv. 7:5603–5607. 2023. View Article : Google Scholar : PubMed/NCBI

30 

Teachey DT, Lacey SF, Shaw PA, Melenhorst JJ, Maude SL, Frey N, Pequignot E, Gonzalez VE, Chen F, Finklestein J, 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. View Article : Google Scholar : PubMed/NCBI

31 

Al-Salama ZT: Emapalumab: First global approval. Drugs. 79:99–103. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2025
Volume 29 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cai W, Lu Y, He H, Li J, Liu S, Geng H, Yang Q, Zeng L, Wu D, Li C, Li C, et al: Efficacy of emapalumab in the management of anti‑CD19 chimeric antigen receptor T‑cell therapy‑associated cytokine release syndrome: A report of two cases. Oncol Lett 29: 71, 2025.
APA
Cai, W., Lu, Y., He, H., Li, J., Liu, S., Geng, H. ... Li, C. (2025). Efficacy of emapalumab in the management of anti‑CD19 chimeric antigen receptor T‑cell therapy‑associated cytokine release syndrome: A report of two cases. Oncology Letters, 29, 71. https://doi.org/10.3892/ol.2024.14817
MLA
Cai, W., Lu, Y., He, H., Li, J., Liu, S., Geng, H., Yang, Q., Zeng, L., Wu, D., Li, C."Efficacy of emapalumab in the management of anti‑CD19 chimeric antigen receptor T‑cell therapy‑associated cytokine release syndrome: A report of two cases". Oncology Letters 29.2 (2025): 71.
Chicago
Cai, W., Lu, Y., He, H., Li, J., Liu, S., Geng, H., Yang, Q., Zeng, L., Wu, D., Li, C."Efficacy of emapalumab in the management of anti‑CD19 chimeric antigen receptor T‑cell therapy‑associated cytokine release syndrome: A report of two cases". Oncology Letters 29, no. 2 (2025): 71. https://doi.org/10.3892/ol.2024.14817