ISSN: 2576-3881

Journal of Cytokine Biology
Open Access

Like us on:

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)
  • Mini Review   
  • J Cytokine Biol 8:450, Vol 8(4)
  • DOI: 10.4172/2576-3881.1000450

A review of an explanation of Cytokine Release Syndrome

Chen Yung*
Department of Pharmacology and Toxicology, College of Pharmacy, King Saudia University, Riyadhanalya, Saudi Arabia
*Corresponding Author: Chen Yung, Department of Pharmacology and Toxicology, College of Pharmacy, King Saudia University, Riyadhanalya, Saudi Arabia, Email: chen.y@ung.com

Received: 04-Jul-2023 / Manuscript No. jcb-23-105937 / Editor assigned: 06-Jul-2023 / PreQC No. jcb-23-105937 (PQ) / Reviewed: 20-Jul-2023 / QC No. jcb-23-105937 / Revised: 24-Jul-2023 / Manuscript No. jcb-23-105937 (R) / Published Date: 31-Jul-2023 DOI: 10.4172/2576-3881.1000450

Abstract

Cancer immunotherapy aims to eliminate cancerous tissues by using the immune system’s capabilities. After decades of study, a variety of cancer immunotherapies have demonstrated unambiguous clinical efficacy. These include graft-versus-leukemia, which eliminates leukaemia after hematopoietic stem cell transplantation (HSCT), monoclonal antibodies (mAbs), which prolong survival for people with B-cell lymphomas and breast cancer that expresses the HER2 gene, and a therapeutic cancer vaccine for hormone-refractory prostate cancer. Bispecific mAbs have mediated impressive responses in B-cell acute lymphoblastic leukemia7 (ALL), and dramatic antitumor effects have been observed using adoptive T-cell immunotherapy, which is increasingly using genetic engineering to create tumour antigen-specific T cells. Recently, mAbs that block key checkpoints on T cells have improved survival in metastatic melanoma and induced antitumor effects in other cancers.

Keywords

Cancer immunotherapy; T cells; Lymphoblastic; Hematopoietic; Leukaemia

Introduction

Adults who have relapsed and refractory (r/r) acute lymphoblastic leukaemia (ALL) typically have a terrible prognosis and pass away from their illness. This setting has contributed to the enthusiasm surrounding the results obtained with the T cell engagers blinatumomab (a bispecific antibody that guides effector T cells to B cells with its anti-CD3 and anti-CD19 arms) and chimeric antigen receptor (CAR) T cells modified to target CD19) [1]. Both substances function by directing cytotoxic T cells towards the CD19 antigen produced on cancerous B cells, thereby realising the anti-tumor potential of a cell-mediated immune response. Adult and paediatric patients with r/r ALL who received 2nd generation anti-CD19 CAR T cells experienced complete remission (CR) rates of 67%-93%. In a significant multicenter study involving 189 patients, blinatumomab single-agent therapy resulted in a 43% CR rate [2].

Highly refractory B-cell malignancies have responded admirably to T cell-engaging treatments, but managing toxicity is a particular difficulty with this immunotherapy. T cell-engaging therapies harness the cell-mediated immune response and guide it against cancerous cells without using the major histocompatibility complex by connecting target antigen-expressing malignant cells with deadly T cells. In earlyphase clinical trials for relapsed/refractory B-cell leukaemias and lymphomas, 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, have both demonstrated striking responses.1–5 However, CRs have been linked to cytokine release syndrome (CRS), which can range in severity from mild to fatal. These trials have shown significant complete response (CR) rates [3].

Pathophysiology:

The type of triggers causing asthma exacerbations is probably one of the main determinants of the pathophysiology of WEA. No evidence exists to suggest that the pathophysiology of WEA differs from the pathophysiology of asthma exacerbations seen in NWRA when the triggers are common allergens. It is believed that the pathophysiology mirrors that of IIA when the triggers are irritating substances. As proven in New York firefighters who assisted on September 11, 2001, an airway epithelium injury is expected to play a crucial role, and the severity of the epithelial layer injury may be connected with the impairment of respiratory function.118 Oxidative stress is a major factor in the pathophysiology of chlorine exposure in mouse models, and administering antioxidants can lessen epithelium damage.119 The degree of harm is expected to depend on the dosage of chlorine exposure, as seen in the animal models of exposure. An increase in the parallel transport activity of pendrin and NDBCE in these patients may be the pathogenesis. This could be the aetiology of what was formerly thought to be a “chloride shunt disorder.” Additionally, these patients will exhibit hypooreninemic hypoaldosteronism, which includes extended EABV, suppressed PRenin, and elevated PAldosterone. When the carbonic anhydrase inhibitor acetazolamide is administered to patients with this disease, it is anticipated that they will excrete more K+ ions and that HCO3 ions will be delivered to the CDN at a higher rate. Those who have hyporeninemic hypoaldosteronism and diabetic nephropathy are two examples of this pathology [4].

CAR T-cell treatment for leukaemia

Despite its considerable effectiveness, CAR T-cell therapy administration is usually made more difficult by the leukaemia cells’ poor clearance rate in immune-privileged locations and the emergence of serious adverse events (AEs). Due to worries about poor response and treatment-related neurotoxicity, CNSL has rarely been the subject of any clinical trials using CAR T-cell therapy. Furthermore, the majority of CAR T-cell trials often have an exclusion criterion for patients with severe CNS illness and active neurologic symptoms. CR with manageable and reversible neurotoxicity was obtained in a patient with resistant secondary CNS diffuse large B-cell lymphoma in 2017, according to a report that confirmed the efficient trafficking of CAR T cells into the CNS. Patients with R/R B-ALL with CNSL may benefit from CAR T-cell therapy, which has an acceptable safety profile, according to a small number of clinical reports that describe instances of IV-delivered CD19 CAR T cells mediating leukaemia cell clearance from the cerebrospinal fluid (CSF) with fully reversible toxicity [5]. To our knowledge, CAR T-cell treatment for CNSL has not yet been the subject of any extensive, specialised case series reporting on response assessment or long-term survival. Additionally, it is necessary to assess the frequency and severity of CAR T-cell therapy-related adverse events (AEs), particularly neurotoxicity, in a larger group of patients with CNS involvement. We discuss the analysis of CD19-specific CAR T cell-based therapy’s efficacy, tolerability, and feasibility in patients with R/R B-ALL with CNSL [6].

Profile of CRS’s Cytokines:

A cure for many haematological illnesses is allogeneic stem cell transplantation (allo-SCT). Some transplant-related problems, like graft-versus-host disease (GVHD), may be caused by the emergence of a so-called “cytokine storm” in the early stages, particularly during the conditioning regimen. The training programme promotes tissue damage and the release of inflammatory cytokines in experimental animal models, which starts an inflammatory response. The release of cytokines during conditioning may also contribute to the development of GVHD in a clinical situation. The effect of the cytokine profile during conditioning on transplant outcomes is still unknown because there are few research that have examined the cytokine profiles in individuals undergoing allo-SCT, particularly haploidentical stem cell transplantation (haplo-SCT) [7].

Risk Elements for CRS:

1. Increasing clinical experience with CRS and infusion reactions.

2. Definition and Intensity of CRS.

3. Alignment on Defining and Grading CRS.

4. Method for Measuring CRS during a Clinical Development Programme.

Materials and Methods

Cytokine Release Syndrome (CRS) is a potentially life-threatening condition characterized by an excessive release of cytokines, which are small signaling proteins involved in immune responses. CRS is most commonly associated with certain immunotherapies, such as chimeric antigen receptor (CAR) T-cell therapy, and can also occur in the context of infections or autoimmune diseases. The materials and methods used to study and manage CRS depend on the specific circumstances and severity of the syndrome. Here are some general aspects:

Clinical Assessment: The first step in managing CRS is a thorough clinical assessment of the patient. This includes monitoring vital signs, assessing organ function, and evaluating symptoms such as fever, hypotension, respiratory distress, and neurologic changes. Various scoring systems, such as the Penn Grading Scale or the Lee Criteria, may be used to classify the severity of CRS [8].

Laboratory Investigations: Blood samples are collected for laboratory investigations, which can help in the diagnosis and monitoring of CRS. These tests may include complete blood count, liver function tests, coagulation profile, and measurement of cytokine levels (e.g., interleukin-6, interferon-gamma, tumor necrosis factoralpha).

Imaging Studies: Imaging techniques like chest X-rays or computed tomography (CT) scans may be performed to assess lung involvement and rule out other causes of respiratory symptoms [9].

Supportive Care: Symptomatic management and supportive care play a crucial role in CRS treatment. This can include measures such as fever control with antipyretics, fluid resuscitation to maintain blood pressure, oxygen therapy, and mechanical ventilation if respiratory distress is severe.

Immunomodulatory Therapies: In severe cases of CRS, immunomodulatory therapies may be required to dampen the excessive immune response. These can include: Systemic corticosteroids like methylprednisolone or dexamethasone are commonly used to suppress inflammation and cytokine production.

Interleukin-6 (IL-6) Inhibitors: Monoclonal antibodies targeting IL-6, such as tocilizumab or siltuximab, can be used to block the effects of IL-6, a key cytokine involved in CRS [10].

Janus Kinase (JAK) Inhibitors: Drugs like ruxolitinib, which inhibit JAK signaling pathways, have shown promise in managing CRS by reducing cytokine production.

Other Therapies: Additional immunomodulatory agents like anakinra (an IL-1 receptor antagonist), etanercept (a tumor necrosis factor inhibitor), or anti-thymocyte globulin (ATG) may be considered in certain cases.

Close Monitoring: Patients with CRS require close monitoring in an intensive care setting. Vital signs, organ function, and laboratory parameters are frequently assessed to guide the management and adjust treatments accordingly. CRS can vary depending on the individual patient’s condition, the underlying cause of CRS, and the healthcare provider’s judgment. Treatment decisions should be made in consultation with a qualified healthcare professional experienced in managing CRS or related conditions.

Result

The interpretation of these results would require the expertise of a healthcare professional who can assess the patient’s overall clinical condition, symptoms, and medical history. They would consider the laboratory results in conjunction with other clinical findings to make an accurate diagnosis and determine the appropriate management plan for CRS. If you have specific laboratory results or data that you would like assistance with, please provide more details, and I’ll do my best to provide relevant information or explanations [11].

Conclusion

In conclusion, cytokine release syndrome (CRS) is a complex condition characterized by an excessive release of cytokines, leading to systemic inflammation and potentially severe symptoms. The materials and methods employed in the study and management of CRS involve clinical assessment, laboratory investigations, imaging studies, supportive care, and immunomodulatory therapies. Clinical assessment includes monitoring vital signs, evaluating organ function, and assessing symptoms. Laboratory investigations may involve blood tests to evaluate complete blood counts, liver function, coagulation profiles, and cytokine levels. Imaging studies such as X-rays or CT scans can help assess lung involvement. Supportive care measures focus on managing symptoms and maintaining organ function. Immunomodulatory therapies, such as corticosteroids, IL-6 inhibitors, JAK inhibitors, and other agents, may be used to dampen the immune response [12].

Close monitoring of the patient’s condition is crucial, often requiring intensive care and regular assessment of vital signs and laboratory parameters. It’s important to note that the specific materials and methods used may vary depending on the patient’s individual circumstances and the healthcare provider’s judgment. It’s always recommended to consult with a qualified healthcare professional experienced in managing CRS or related conditions for an accurate diagnosis, personalized treatment plan, and interpretation of specific laboratory results.

Acknowledgement

None

References

  1. Hale G, Clark M, Marcus R, Winter G, Dyer MJS (1988) Remission induction in non-Hodgkin, lymphoma with reshaped human monoclonal antibody CAMPATH 1-H. Lancet 2:1394-1399.
  2. Indexed at, Google Scholar, Crossref

  3. Hale G, Xia M-Q, Tighe HP, Dyer MJS, Waldmann H (1990) The CAMPATH-1 antigen (CDw52). Tissue Antigens 35:118-127.
  4. Indexed at, Google Scholar, Crossref

  5. Schumacher TN, Heemels MT, Neefjes JJ, Kast WM, Melief CJ, et al. (1990) Direct binding of peptide to empty MHC class I molecules on intact cells and in vitro. Cell 62:563-567.
  6. Indexed at, Google Scholar, Crossref

  7. Kelly A, Powis SH, Kerr LA, Mockridge I, Elliott T, et al. (1992) Assembly and function of the two ABC transporter proteins encoded in the human major histocompatibility complex. Nature 355:641-644.
  8. Indexed at, Google Scholar, Crossref

  9. Isaacs JD, Watts R, Hazleman BL, Hale G, Keogan MT, et al. (1992). Humanised monoclonal antibody therapy for rheumatoid arthritis. Lancet 340:748-752.
  10. Indexed at, Google Scholar, Crossref

  11. Pachlopnik Schmid J, Canioni D, Moshous D, Touzot F, Mahlaoui N, et al. (2011) Clinical similarities and differences of patients with X-linked lymphoproliferative syndrome type 1 (XLP-1/SAP deficiency) versus type 2 (XLP-2/XIAP deficiency). Blood 117:1522-1529.
  12. Indexed at, Google Scholar, Crossref

  13. Henter JI, Horne A, Arico M, Egeler RM, Filipovich AH, et al. (2007) HLH-2004: diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer 48:124-131.
  14. Indexed at, Google Scholar, Crossref

  15. Zhang K, Jordan MB, Marsh RA, Johnson JA, Kissell D, et al. (2011) Hypomorphic mutations in PRF1, MUNC13-4, and STXBP2 are associated with adult-onset familial HLH. Blood 118:5794-5798.
  16. Indexed at, Google Scholar, Crossref

  17. Mathieson PW, Cobbold SP, Hale G, Clark MR, Oliveira DBG (1990) Monoclonal antibody therapy in systemic vasculitis. N Eng J Med 323:250-254.
  18. Indexed at, Google Scholar, Crossref

  19. Zhou P, Yang X, Wang X, Zhang L, Zhang, et al. (2020) A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 579:270-273.
  20. Indexed at, Google Scholar, Crossref

  21. Padmanabhan, Connelly-Smith L, Aqui N (2019) Guidelines on the use of therapeutic apheresis in clinical practice- evidence-based approach from the writing committee of the American Society for Apheresis: the eighth special issue. J Chromatogr 34:167-354.
  22. Indexed at, Google Scholar, Crossref

  23. Marsh RA, Madden L, Kitchen BJ, Mody R, McClimon B, et al. (2010) XIAP deficiency: a unique primary immunodeficiency best classified as X-linked familial hemophagocytic lymphohistiocytosis and not as X-linked lymphoproliferative disease. Blood 116:1079-1082.
  24. Indexed at, Google Scholar, Crossref

Citation: Yung C (2023) A review of an explanation of Cytokine Release Syndrome.J Cytokine Biol 8: 450. DOI: 10.4172/2576-3881.1000450

Copyright: © 2023 Yung C. This is an open-access article distributed under theterms of the Creative Commons Attribution License, which permits unrestricteduse, distribution, and reproduction in any medium, provided the original author andsource are credited.

Top