Next Article in Journal
Diagnostic and Prognostic Value of CEA and CA19-9 in Colorectal Cancer
Previous Article in Journal
Osteocalcin, Osteopontin and RUNX2 Expression in Patients’ Leucocytes with Arteriosclerosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cardiotoxicity Associated with Anti-CD19 Chimeric Antigen Receptor T-Cell (CAR-T) Therapy: Recognition, Risk Factors, and Management

1
Houston Methodist Cancer Center, Houston Methodist Hospital, 6445 Main Street, Outpatient Center, 24th Floor, Houston, TX 77030, USA
2
Houston Methodist DeBakey Heart and Vascular Center, 6565 Fannin St, Houston, TX 77030, USA
3
Callahan Cancer Center, Great Plains Health, 601 W Leota St, North Platte, NE 69101, USA
*
Authors to whom correspondence should be addressed.
Submission received: 9 February 2021 / Revised: 4 March 2021 / Accepted: 15 March 2021 / Published: 17 March 2021

Abstract

:
Chimeric antigen receptor T-cells (CAR-T) are improving outcomes in pediatric and adult patients with relapsed or refractory B-cell acute lymphoblastic leukemias and subtypes of non-Hodgkin Lymphoma. As this treatment is being increasingly utilized, a better understanding of the unique toxicities associated with this therapy is warranted. While there is growing knowledge on the diagnosis and treatment of cytokine release syndrome (CRS), relatively little is known about the associated cardiac events that occur with CRS that may result in prolonged length of hospital stay, admission to the intensive care unit for pressor support, or cardiac death. This review focuses on the various manifestations of cardiotoxicity, potential risk factors, real world and clinical trial data on prevalence of reported cardiotoxicity events, and treatment recommendations.

1. Introduction

Chimeric antigen receptors (CAR) are synthetic immunoreceptors comprised of a single-chain variable fragment acquired from an immunoglobulin with an affinity directed toward a specific tumor antigen, and an intracellular signaling moiety connected by transmembrane domains [1,2,3]. The genetic sequencing is introduced to a patient’s T-lymphocytes ex vivo via lentivirus or non-viral vectors, which are than expanded and returned to the circulation of a patient that has received lymphodepleting chemotherapy, commonly fludarabine and cyclophosphamide [4,5]. Once the modified cells are introduced to the patient’s circulation, the CAR-T cells can preferentially target tumor cells with aberrant expression of a specific antigen (Figure 1) [6].
The development of genetically modified CAR-T cells targeting CD19, an antigen that is frequently over-expressed in various B-cell malignancies, is changing the therapeutic landscape in pediatric and adult patients with relapsed/refractory (r/r) B-cell acute lymphoblastic leukemia (B-ALL) and subtypes of non-Hodgkin Lymphoma (NHL). Following the results of the study of Efficacy and Safety of CTL019 in Adult DLBCL Patients (JULIET trial) and study of Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia (ELIANA trial), Tisagenlecleucel (CTL019; Novartis Pharmaceuticals) was approved by the food and drug administration (FDA) for B-ALL in 2017 and large B-cell lymphoma (LBCL) (including diffuse large B-cell lymphoma [DLBCL], high-grade B-cell lymphoma [BCL], and DLBCL arising from follicular lymphoma [FL]) in 2018. These approvals encompass pediatric and young adult patients up to the age of 25 years with second or later disease relapse or that are refractory to two or more lines of systemic therapy [7,8]. Following the results from the safety and efficacy of KTE-C19 in adults with refractory aggressive non-Hodgkin lymphoma (ZUMA-1 trial), Axicabtagene Ciloleucel (KTE-C19; Kite Pharma Incorporated) was approved by the FDA in 2017 in adults with LBCLs (DLBCL, primary mediastinal large B-cell lymphoma [PML], high-grade BCL, and DLBCL arising from FL) that are relapsed or refractory to two or more lines of systemic therapy [9]. In 2020, following the study on KTE-X19 CAR T cell therapy in relapsed or refractory mantle-cell lymphoma (ZUMA-2 trial), Brexucabtagene Autoleucel (KTE-X19, Kite Pharma Incorporated) was approved by the FDA for the treatment of adult patients with r/r mantle cell lymphoma (MCL) [10].
While these agents are reporting unparalleled response rates ranging from 50–93% in the r/r setting [7,8,9,10], these therapies carry unique and potentially significant toxicities. In clinical trials and retrospective institutional studies, cytokine release syndrome (CRS) is a frequently encountered toxicity. While there is a growing body of literature aimed at studying and ameliorating the potential impacts of these toxicities following CAR-T administration, there is little data on cardiotoxic events. While uncommonly reported in the pivotal trials [11], as the treatment population receiving CD19 CAR-T grows, understanding potential cardiac events that may occur, predisposing risk factors, and management recommendations is necessary to improve non-relapse related outcomes. The following review aims to provide a comprehensive review on cardiac toxicities, as well as diagnostic and treatment considerations for pediatric and adult patients undergoing CD19 CAR-T infusions.

2. Cardiotoxicity

Cardiac events following CD19 CAR-T infusion frequently occur as a complication arising in patients with grade 3–4 CRS (Table 1) [12,13,14,15]. Upon recognition of the tumor antigen, CAR-T cells release pro-inflammatory cytokines including interferon gamma (IFNγ), interleukin (IL)-1, IL-2 receptor alpha (RA), tumor necrosis factor alpha (TNFα), and IL-6 to induce a cytotoxic response [15]. The release of these cytokines, particularly IL-6, also plays a role in the pathogenesis of CRS, including the recruitment of additional T-lymphocytes [16,17]. High levels of circulating IL-6 can also lead to myocardial stunning which may be clinically indistinguishable from septic cardiomyopathy [18,19]. Furthermore, CRS results in the activation of prostaglandins, which can also impart a risk of cardiotoxic events, such as tachycardia and hypotension [15,16]. It is also possible that CAR-T results in a direct or “off-target” cardiotoxic injury as a result of cross-reactivity between T-cells and Titin, a reaction observed in an early CAR-T targeting melanoma-associated antigen 3 (MAGEA3) [15,18,20].
A wide array of cardiac events has been reported in both pediatric and adult patients, including sinus tachycardia, arrhythmias, ST segment changes on the electrocardiogram (ECG), left ventricular systolic dysfunction, decompensated heart failure, life-threatening hypotension requiring vasopressor support, and rarely, cardiac death (Table 2). In the pivotal trials, patients with pre-existing or recent cardiovascular events were excluded from enrollment (Table 3), but many patients included in these trials had previously received therapies that increase the risk of cardiac disease, including treatment with anthracycline containing treatment regimens, irradiation, and allogenic stem cell transplantation. Outside of the clinical trials, data reporting on cardiotoxic events are starting to accumulate. Currently, cardiac disease is not an absolute contraindication to proceeding with CAR-T [21,22,23].

2.1. Cardiotoxicity in Pediatric and Young Adult Patients

Hypotension is frequently reported in pediatric patients and is often a complication of CRS. In the ELIANA trial, 29% of patients developed hypotension, and 17% were grade 3–4 (Table 1 and Table 4) [8]. Other studies of pediatric and young adult patients have found that 18–38% of patients will develop grade 3–4 hypotension (Table 5) [8,24,25,26,27], with 13–27% of these patients requiring vasopressor support [24,25,28,29], and up to one-third of patients requiring admission to the intensive care unit (ICU) [25]. In a retrospective study by Fitzgerald et al., 13 patients that developed cardiovascular toxicity developed fluid-refractory shock that necessitated the use of alpha agonists. Ten of these patients required additional vasoactive substances. In this cohort, cardiovascular dysfunction developed at a median of 5 days after the infusion and lasted a median of 4 days. All patients had a preceding fever prior to the onset of shock [30].
A wide array of other cardiac events is reported in the pediatric and young adult population, frequently resulting as a sequela of hypotension and CRS (Table 5). In the ELIANA trial, 3 (4%) patients developed left ventricular dysfunction, 2 (2.7%) patients developed cardiac failure, and 3 (4%) had cardiac arrest (Table 4) [8]. In a retrospective study by Burstein et al., 10 (41%) of the 24 patients that developed hypotension requiring inotropic support went on to develop systolic dysfunction, and one patient had cardiac arrest. Six of these patients required milrinone, and 5 of these patients required additional inotropic support including dopamine, epinephrine, norepinephrine, or vasopressin. Six of these patients had ST segment changes. All patients had grade 3–4 CRS. [29]. In a retrospective study of pediatric patients that developed cardiotoxicity by Shalabi et al., 6 (16%) patients with CRS developed cardiac dysfunction, and all required monitoring in the intensive care unit. No cardiotoxicity was reported in the patients that did not develop CRS. Patients with cardiac dysfunction in this study were more likely to have earlier CRS, more severe CRS, receive tocilizumab, and have a lower baseline global longitudinal strain (GLS). In this patient cohort, of the 37 patients that developed CRS, 9 (24%) developed hypotension requiring vasopressors, with 3 of these patients requiring two or more vasopressor agents [28].
In children, cardiotoxic complications associated with CAR-T appear to largely be self-limited, with most patients recovering cardiac function back to pre-treatment baseline, including patients that had cardiac arrest [28,29]. In the study by Shalabi et al, all 6 individuals with heart failure had recovery by 3 months, and in the retrospective study by Burstein, 6 of the 7 with impaired systolic function had recovery by 6 months of follow up. None of the cardiac events contributed to mortality [28,29].

2.2. Cardiotoxicity in Adult Patients

In the JULIET, ZUMA-1, and ZUMA-2 trials assessing CD19-CAR-T cells in adults, hypotension ranged from 26–59%, and hypotension or shock requiring pressor support occurred in 9–22% of patients. Tachycardia was also reported between 11–39% of patients, but cardiac arrest and heart failure were not reported (Table 4) [7,9,10]. Outside of the pivotal trials, the first retrospective study assessing cardiovascular outcomes came from Alvi et al. [31]. In this analysis involving patients treated with Axicabtagene Ciloleucel, Tisagenlecleucel, and investigational CD19 CAR-T, 17 (12%) developed cardiovascular events with a median time of 21 days to the event, and a median follow up of 10 months. There were 6 cardiac deaths, 6 accounts of acute heart failure, and 5 occurrences of new-onset arrhythmias (Table 6). There appeared to be a close relationship between cardiac events, cardiac injury, and CRS; all patients had > grade 2 CRS, and 16 patients with cardiotoxicity had a positive troponin out of a total of 29 patients in the entire cohort with a positive troponin [31].
In a recent study assessing major cardiac events (MACE) including cardiovascular death, symptomatic heart failure, acute coronary syndrome, ischemic stroke, and de novo cardiac arrhythmia in patients that received CAR-T, Lefebvre found that 31 (21.4%) patients developed 41 cardiac events within a median of 11 days, and over a follow-up period of 753 days. Kaplan–Meier methodology found that the cumulative incidence for a major cardiac event was 17% at 30 days, 19% at 6 months, and 21% at 12 months following CD19-CAR-T infusion, suggestive of a longer-term risk of cardiovascular events [32]. In another study conducted by investigators at the Dana Farber Cancer institute, the incidence of cardiomyopathy was assessed in 187 patients receiving CD19 CAR-T for NHL. In this study 155 (83%) patients developed ≥2 CRS, and there were 116 with serial echocardiograms available for evaluation. Of these 116, 12 (10.3%) developed new (11) or progressive (1) cardiomyopathy within a median of 12.5 (2–24) days following CAR-T infusion. Of these, 11 had ≥2 CRS, and all were treated with Tocilizumab. In addition, 5 (42%) required vasopressor support. Of these 12, the ejection fraction improved in 9 of the 12 patients over 168.5 (3–471) days, with normalization 6 and partial recovery in 3. The 3 without recovery in cardiac function died [18].
Contrary to what is seen in the pediatric and young adult patients that develop cardiotoxicity, adult patients receiving CD19 CAR-T agents that develop cardiotoxicity do not always have resolution of their cardiac events, and in some cases, have fatal events. While there are risk factors becoming recognized for patients that may be at risk of developing CRS and association with cardiotoxicity, there needs to be a standardized pre-treatment protocol for screening these patients with recommendations on how to proceed for patients with increasingly recognized risks for cardiotoxicity.

3. Risks for Developing Cardiotoxic Events

Cardiotoxicity is frequently a sequela of CRS, so understanding risks that may predispose to CRS should be considered in patients, especially those with pre-existing cardiovascular disease. Patients that are at risk of developing CRS are patients with high disease burden, an older age at the time of infusion, higher-intensity lymphodepleting regimen, utilization of fludarabine and cyclophosphamide during lymphodepleting chemotherapy, higher infused CAR T-cell doses, use of unselected bulk CD8+ T cells, high levels of CTL019+ CD8 and CD3 cells, the presence of inflammatory markers including a higher peak of C-reactive protein, and severe thrombocytopenia [25,27,33,34] (Table 7).
Knowledge or risk factors directly contributing to cardiotoxicity are largely limited to retrospective studies. In pediatric and young adult patients, the risk of developing hypotension requiring inotropic support appears to be associated with a pre-treatment blast percentage >25% on bone marrow biopsy, lower baseline ejection fraction or GLS, or pre-existing diastolic dysfunction [28,29]. Studies have not found previous anthracycline-based chemotherapy, radiation exposure, or history of stem cell transplantation to be associated with the risk of developing clinically significant hypotension or cardiac dysfunction [16,29] (Table 7).
In adults, Alvi et al. reported that concomitant CRS, troponin elevation, and the time of onset of elevated troponin in CRS to the first administration of tocilizumab, are associated with an increased risk of developing a cardiovascular event [31]. Lefebvre et al. found that patients at risk for developing MACE were older, had a higher prevalence of baseline cardiovascular risk factors at baseline, higher baseline creatinine, and grade 3–4 CRS [32]. In addition, prior, aspirin use, statin use, and insulin use had a higher association with MACE [32]. The subtype of malignancy did not seem to impart a risk of cardiotoxicity [32]. Ganatra et al. found that adults that were older, had high-grade CRS, hyperlipidemia, known coronary artery disease, or use of renin-angiotensin inhibitors and beta-blockers at baseline had a higher risk of developing post-CAR-T cardiomyopathy [18] (Table 7).

4. Management and Treatment

4.1. Pre-CAR-T Infusion Cardiovascular Considerations

Patients and pertinent risk factors that may predispose to cardiotoxicity should be identified prior to initiation of CAR-T infusion. In retrospective studies, the timing from CAR-T infusion to reported cardiac event ranged from a median of 5–21 days [18,28,29,30,31,32]. While baseline cardiac evaluation is typically institution dependent, the American Council of Cardio-Oncology recommends the consideration of a baseline ECG and echocardiographic assessment of cardiac function. Specifically, the presence of baseline arrhythmias, structural heart disease, or coronary artery disease (CAD) should be assessed [35]. If a patient is found to have systolic or diastolic dysfunction, valvular disease, cardiomyopathy, CAD, or pre-existing risk factors, then they should be referred for evaluation by a Cardio-Oncologist for further risk stratification and optimization of cardiovascular function [35,36]. Ischemic burden should be assessed with stress test imaging in patients with known CAD or risk factors that may predispose to cardiovascular decompensation [35,36]. Prior to CAR-T infusion, patients with pre-existing systolic and diastolic dysfunction should be volume optimized and monitored closely during infusion due to the risk for fluid shifts [35]. In addition, outpatient medications should be reviewed, particularly antihypertensives, antiplatelets, and other anticoagulants given the risk of hemodynamic compromise after CAR-T infusion as well as cytopenias due to lymphodepletion (Figure 2) [35,37].

4.2. Clinical Monitoring during and after CAR-T Infusion

Patients that develop ≥grade 2 CRS are at a higher risk of developing cardiotoxicity, so close clinical monitoring for the development of CRS is crucial. Although recommendations vary, monitoring may include blood pressure checks, 12-lead ECGs, telemetry, and cardiac biomarker assessments including cardiac troponin and brain natriuretic peptide in patients that are showing clinical evidence of CRS [35,36,38]. If hypotension or tachycardia develops, patients should undergo volume resuscitation. However, close attention should be paid to symptoms of vascular leak and pulmonary edema [37]. If refractory to fluid boluses, transfer to the ICU for close hemodynamic monitoring and vasopressor initiation should be considered. Reassessment of left ventricular function with an echocardiogram can help elucidate a cardiogenic component to shock [35] (Figure 3).
Although troponemia has been associated with an increased risk of developing cardiovascular events after CAR-T infusions [31], there is limited data on the benefit of obtaining routine biomarker assessments. While additional studies need to be done in both pediatric and adult patients to corroborate this, it should be considered particularly in those that have known pre-existing cardiovascular disease.

4.3. Management of Cardiovascular Events

There are currently no formally accepted guidelines for the management of CAR-T induced cardiotoxicity. Data for specific cardiovascular interventions are lacking; however, treatment recommendations largely revolves around supportive care, including hemodynamic support and management of the precipitating CRS (Figure 3).

4.3.1. Supportive Care

Supportive care must be initiated in patient with hypotension, myocardial depression, arrhythmias, and sinus tachycardia. In hypotension, initial treatment with intravenous crystalloids is recommended [35]. Close clinical monitoring to diagnose refractory hypotension and acute fluid shifts predisposing to capillary leak and respiratory failure is important [37]. Patients with refractory hypotension should be initiated on vasopressor support and monitored closely in the ICU. If there is associated tachycardia and fever, appropriate infectious workup and broad-spectrum antimicrobial agents should be initiated [39]. In the setting of depressed myocardial dysfunction or cardiogenic shock, inotropic support with agents like dobutamine can also be considered [36].

4.3.2. IL-6 Inhibitor Therapy

CRS is a systemic inflammatory response mediated by the release of cytokines such as IL-6, IL-10, interferon gamma and tumor necrosis factor alpha. Once secreted, IL-6 has pro-inflammatory effects which play a substantial role in the pathogenesis of CRS including capillary leak, hypotension, complement activation and myocardial dysfunction [40,41]. Tocilizumab is a monoclonal antibody that inhibits the binding of IL-6 to its receptor. It acts on both membrane bound and soluble IL-6 receptors thereby inhibiting downstream IL-6 signaling [40]. It was approved by the FDA for use in severe or life-threatening CAR-T mediated CRS in adult and pediatric patients older than 2 years alone or in combination with steroids, following the results of a retrospective analysis of pooled data from 5 clinical trials [42].
The management of CRS has important implications in cardiotoxicity events. Alvi et al. reported that in patients with troponemia following CAR-T infusion, the risk of a cardiovascular event increases with each 12-h delay in administration of tocilizumab [31]. It is generally accepted to start tocilizumab in the presence of hemodynamic instability requiring vasopressor support, increasing oxygen requirement and/or evidence of end-organ dysfunction (unstable arrhythmia, myocardial infarction, cardiomyopathy) [35,37,43].
An alternative IL-6 inhibitor, siltuximab, has also been used in CRS. Its mechanism differs to that of tocilizumab in that is directly binds to circulating IL-6 [37,40]. Prior studies have administered it either interchangeably with tocilizumab or as an option in refractory CRS [38]. At this time, it is not FDA approved and its use is investigational, however, recommendations are to consider it in patients with CRS that have not responded to tocilizumab and/or corticosteroids [35,41,44]. It is not known if expedient administration of siltuximab would similarly reduce the risk of developing cardiac events in patients with troponemia, like what is seen with tocilizumab [31].

4.3.3. Corticosteroid Therapy

The use of corticosteroids has proven to be effective in CRS, particularly when it is severe and refractory to other interventions [36,44]. Although practices vary, it is commonly considered a 2nd line agent. However, it may also be administered in conjunction with tocilizumab in cases of severe CRS [37,38,40].
Further research is needed to define the role and safety of corticosteroids for treatment of cardiotoxicity events related to, or independent of CRS. “Stress dose” corticosteroids are frequently used in cases of refractory hypotension stemming from septic shock, and in other cases in which the hypothalamic-pituitary access (HPA) may be significantly impaired [45,46]. However, in the setting of immune activation, pro-inflammatory cytokines such as IL-6 are known to augment the HPA axis and increase the level of circulating corticosteroids [47]. While administration of systemic corticosteroids does not impart a detrimental impact on CAR-T efficacy, its use to treat or prevent cardiac events is poorly understood. It is becoming routinely used in CRS, but in a study of pediatric patients, administration of steroids did not reduce the risk of developing cardiac dysfunction (p = 0.11) [28]. Additional data related to corticosteroid use and cardiotoxicity remain sparse, and additional studies are needed to better understand the therapeutic applications.

4.3.4. The Role of IL-1 Therapy

There are ongoing studies exploring the use of other agents in CRS management. IL-1 is a particularly enticing target given its pro-inflammatory function, pathophysiologic role in CRS, and its role in cardiovascular disease [48]. Phase II and III trials have shown that blocking IL-1 may prevent recurrent atherothrombotic cardiovascular events, increase exercise capacity in heart failure patients and prevent heart failure following a myocardial infarction [48]. Anakinra is an IL-1 inhibitor that is FDA approved for rheumatoid arthritis. However, pre-clinical data have indicated a potential role in CRS [40,41]. Two animal model studies have reported that IL-1 blockade can prevent CRS, thereby reducing mortality [49,50]. A case report of two patients using tocilizumab and anakinra (200 mg subcutaneous three times daily) in a patient treated with anti-BCMA CAR T was published recently [51]. Only one dose of tocilizumab was given and a taper of anakinra over the following 10 days was done. The authors noticed significant improvement of the patient’s symptoms shortly after anakinra was initiated. They theorize that IL-1 blockage decreased the duration and severity of the CRS as well as the need for further tocilizumab doses [51]. In addition, a phase II clinical trial to explore the role of anakinra further is underway (NCT04359784).

4.3.5. The Role of TNF-α Therapy

TNF-α is a pro-inflammatory cytokine that is elevated following administration of CD19 CAR-T therapy [52]. Therefore, it is possible that subsets of patients with CRS may benefit from anti-TNFα monoclonal antibodies infliximab or soluble TNFα receptor, etanercept. There have been rare reports of the utilization of TNF-α inhibitor therapy for CRS arising from CAR-T therapy. Lee et al. reported on a case of grade 3 CRS arising in a 19-year-old female being treated with CAR-T for EBV-associated lymphoma [13]. Within hours of receiving treatment with etanercept, methylprednisolone, dopamine, and norepinephrine, her symptoms resolved [13]. Furthermore, in a small study of 8 patients with r/r multiple myeloma treated with LCAR-B38M (anti-BCMA CAR-T cells), 3 patients were treated with etanercept (either 25 mg or 50 mg) with excellent clinical response [53]. As the number of clinical trials for CAR-T in MM increase, the role of TNF—α may need to be further explored for severe CRS [54].
Neither of the reports above involved treatment with CD19 CAR-T cells. Whether these patients may derive similar benefit is not currently known. In a report of 2 pediatric patients with pre-B-cell ALL, 1 patient developed severe CRS, which responded to treatment consisting of etanercept, tocilizumab, and corticosteroids [55]. Additional studies are needed to determine if patients receiving CD19 CAR-T cells that develop grade 3 or 4 CRS should be considered to receive anti- TNF-α therapy.

5. Conclusions

As the therapeutic applications of CD19 CAR-T continues to grow in the relapsed/refractory setting of B-ALL and NHL, recognition, and management of associated toxicities in the standard of care setting is crucial. While the unique toxicities associated with CD19 CAR-T such as CRS is well-studied, there remains limited data on identification and management of cardiac toxicities. While this review highlights the currently available literature on risk factors and treatment considerations of cardiotoxicitiy in the pediatric and adult populations, there remains a paucity of guidelines available. Future studies are needed to better direct the management of these patients to reduce the morbidity following CAR-T administration.

Author Contributions

Conceptualization, original draft preparation: E.A.B., C.G., B.T., S.R.P., K.A. Data acquisition and literature review: E.A.B., C.G., B.T., S.R.P., K.A. Writing—review and editing: E.A.B., C.G., B.T., S.R.P., K.A. Supervision: K.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CAR-T: Chimeric antigen receptor T-cell; CRS: Cytokine release syndrome; CAR: Chimeric antigen receptor; r/r: Relapsed/refractory; B-ALL: B-cell acute lymphoblastic leukemia; NHL: non-Hodgkin lymphoma; DLBCL: Diffuse large B-cell lymphoma; BCL: High-grade B-cell lymphoma; LBCL: Large B-cell lymphoma; FL: Follicular lymphoma; PML: Primary mediastinal large B-cell lymphoma; MCL: Mantle cell lymphoma; IFNγ: Interferon-gamma; IL-1: Interleukin-1; IL-2RA; Interleukin-2 receptor alpha; TNFα: Tumor necrosis factor-alpha; IL-6: Interleukin-6; MAGEA3: melanoma-associated antigen 3; ECG: Electrocardiogram; LFT: Liver function tests; MI: Myocardial infarction; PFS: Progression free survival; RFS: Relapse free survival; OS: Overall survival; AE: Adverse events; T-ALL: T-cell acute lymphoblastic leukemia; MACE: Major cardiac event; CLL: Chronic lymphocytic leukemia; GLS: global longitudinal strain; CAD: Coronary artery disease; RAA: Renin-angiotensin-aldosterone; ICU: Intensive care unit; HPY: Hypothalamic-pituitary axis.

References

  1. Lulla, P.D.; Hill, L.C.; Ramos, C.A.; Heslop, H.E. The use of chimeric antigen receptor T cells in patients with non-Hodgkin lym-phoma. Clin. Adv. Hematol. Oncol. 2018, 16, 375–386. [Google Scholar] [PubMed]
  2. Zmievskaya, E.; Valiullina, A.; Ganeeva, I.; Petukhov, A.; Rizvanov, A.; Bulatov, E. Application of CAR-T Cell Therapy beyond Oncology: Autoimmune Diseases and Viral Infections. Biomedcines 2021, 9, 59. [Google Scholar] [CrossRef] [PubMed]
  3. Filin, I.Y.; Solovyeva, V.V.; Kitaeva, K.V.; Rutland, C.S.; Rizvanov, A.A. Current Trends in Cancer Immunotherapy. Biomedcines 2020, 8, 621. [Google Scholar] [CrossRef] [PubMed]
  4. Roberts, Z.J.; Better, M.; Bot, A.; Roberts, M.R.; Ribas, A. Axicabtagene ciloleucel, a first-in-class CAR T cell therapy for aggressive NHL. Leuk. Lymphoma 2017, 59, 1785–1796. [Google Scholar] [CrossRef]
  5. Ghosh, A.K.; Chen, D.H.; Guha, A.; MacKenzie, S.; Walker, J.M.; Roddie, C. CAR T Cell Therapy–Related Cardiovascular Outcomes and Management. JACC Cardio Oncol. 2020, 2, 97–109. [Google Scholar] [CrossRef]
  6. McHayleh, W.; Bedi, P.; Sehgal, R.; Solh, M. Chimeric Antigen Receptor T-Cells: The Future is Now. J. Clin. Med. 2019, 8, 207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Schuster, S.J.; Bishop, M.R.; Tam, C.S.; Waller, E.K.; Borchmann, P.; McGuirk, J.P.; Jäger, U.; Jaglowski, S.; Andreadis, C.; Westin, J.R.; et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2019, 380, 45–56. [Google Scholar] [CrossRef]
  8. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef]
  9. Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef]
  10. Wang, M.; Munoz, J.; Goy, A.; Locke, F.L.; Jacobson, C.A.; Hill, B.T.; Timmerman, J.M.; Holmes, H.; Jaglowski, S.; Flinn, I.W.; et al. KTE-X19 CAR T-Cell Therapy in Relapsed or Refractory Mantle-Cell Lymphoma. N. Engl. J. Med. 2020, 382, 1331–1342. [Google Scholar] [CrossRef]
  11. Penack, O.; Koenecke, C. Complications after CD19+ CAR T-Cell Therapy. Cancers 2020, 12, 3445. [Google Scholar] [CrossRef]
  12. Porter, D.; Frey, N.; Wood, P.A.; Weng, Y.; Grupp, S.A. Grading of cytokine release syndrome associated with the CAR T cell therapy tisagenlecleucel. J. Hematol. Oncol. 2018, 11, 1–12. [Google Scholar] [CrossRef]
  13. Lee, D.W.; Gardner, R.A.; Porter, D.L.; Louis, C.U.; Ahmed, N.; Jensen, M.C.; Grupp, S.A.; Mackall, C.L. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014, 124, 188–195. [Google Scholar] [CrossRef] [Green Version]
  14. Lee, D.W.; Santomasso, B.D.; Locke, F.L.; Ghobadi, A.; Turtle, C.J.; Brudno, J.N.; Maus, M.V.; Park, J.H.; Mead, E.; Pavletic, S.; et al. ASTCT Consensus Grading for Cytokine Release Syndrome and Neurologic Toxicity Associated with Immune Effector Cells. Biol. Blood Marrow Transpl. 2019, 25, 625–638. [Google Scholar] [CrossRef] [Green Version]
  15. Stein-Merlob, A.F.; Rothberg, M.V.; Holman, P.; Yang, E.H. Immunotherapy-Associated Cardiotoxicity of Immune Checkpoint Inhibitors and Chimeric Antigen Receptor T Cell Therapy: Diagnostic and Management Challenges and Strategies. Curr. Cardiol. Rep. 2021, 23, 1–11. [Google Scholar] [CrossRef] [PubMed]
  16. Oved, J.H.; Barrett, D.M.; Teachey, D.T. Cellular therapy: Immune-related complications. Immunol. Rev. 2019, 290, 114–126. [Google Scholar] [CrossRef] [PubMed]
  17. Scheller, J.; Chalaris, A.; Schmidt-Arras, D.; Rose-John, S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim. Biophys. Acta Mol. Cell Res. 2011, 1813, 878–888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Ganatra, S.; Redd, R.; Hayek, S.S.; Parikh, R.; Azam, T.; Yanik, G.A.; Spendley, L.; Nikiforow, S.; Jacobson, C.; Nohria, A. Chimeric Antigen Receptor T-Cell Therapy–Associated Cardiomyopathy in Patients with Refractory or Relapsed Non-Hodgkin Lymphoma. Circulation 2020, 142, 1687–1690. [Google Scholar] [CrossRef]
  19. Pathan, N.; Hemingway, C.A.; Alizadeh, A.A.; Stephens, A.C.; Boldrick, J.C.; Oragui, E.E.; McCabe, C.; Welch, S.B.; Whitney, A.; O’Gara, P.; et al. Role of interleukin 6 in myocardial dysfunction of meningococcal septic shock. Lancet 2004, 363, 203–209. [Google Scholar] [CrossRef] [Green Version]
  20. Linette, G.P.; Stadtmauer, E.A.; Maus, M.V.; Rapoport, A.P.; Levine, B.L.; Emery, L.; Litzky, L.; Bagg, A.; Carreno, B.M.; Cimino, P.J.; et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood 2013, 122, 863–871. [Google Scholar] [CrossRef]
  21. Yáñez, L.; Sánchez-Escamilla, M.; Perales, M.-A. CAR T Cell Toxicity: Current Management and Future Directions. HemaSphere 2019, 3, e186. [Google Scholar] [CrossRef] [PubMed]
  22. Kymriah-Epar-Product-Information. Available online: https://www.ema.europa.eu/documents/product-information/kymriah-epar-product-information_en.pdf (accessed on 1 March 2021).
  23. Yescarta-Epar-Product-Information. Available online: https://www.ema.europa.eu/documents/product-information/yescarta-epar-product-information_en.pdf (accessed on 1 March 2021).
  24. Kochenderfer, J.N.; Somerville, R.P.; Lu, T.; Shi, V.; Bot, A.; Rossi, J.; Xue, A.; Goff, S.L.; Yang, J.C.; Sherry, R.M.; et al. Lymphoma Remissions Caused by Anti-CD19 Chimeric Antigen Receptor T Cells Are Associated with High Serum Interleukin-15 Levels. J. Clin. Oncol. 2017, 35, 1803–1813. [Google Scholar] [CrossRef] [PubMed]
  25. Maude, S.L.; Frey, N.; Shaw, P.A.; Aplenc, R.; Barrett, D.M.; Bunin, N.J.; Chew, A.; Gonzalez, V.E.; Zheng, Z.; Lacey, S.F.; et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 2014, 371, 1507–1517. [Google Scholar] [CrossRef] [Green Version]
  26. Lee, D.W.; Kochenderfer, J.N.; Stetler-Stevenson, M.; Cui, Y.K.; Delbrook, C.; Feldman, S.A.; Fry, T.J.; Orentas, R.; Sabatino, M.; Shah, N.N.; 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 2015, 385, 517–528. [Google Scholar] [CrossRef]
  27. Davila, M.L.; Riviere, I.; Wang, X.; Bartido, S.; Park, J.; Curran, K.; Chung, S.S.; 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. 2014, 6, 224ra25. [Google Scholar] [CrossRef] [Green Version]
  28. Shalabi, H.; Sachdev, V.; Kulshreshtha, A.; Cohen, J.W.; Yates, B.; Rosing, D.R.; Sidenko, S.; Delbrook, C.; Mackall, C.; Wiley, B.; et al. Impact of cytokine release syndrome on cardiac function following CD19 CAR-T cell therapy in children and young adults with hematological malignancies. J. Immunother. Cancer 2020, 8, e001159. [Google Scholar] [CrossRef]
  29. Burstein, D.S.; Maude, S.; Grupp, S.; Griffis, H.; Rossano, J.; Lin, K. Cardiac Profile of Chimeric Antigen Receptor T Cell Therapy in Children: A Single-Institution Experience. Biol. Blood Marrow Transpl. 2018, 24, 1590–1595. [Google Scholar] [CrossRef] [Green Version]
  30. Fitzgerald, J.C.; Weiss, S.L.; Maude, S.L.; Barrett, D.M.; Lacey, S.F.; Melenhorst, J.J.; Shaw, P.; Berg, R.A.; June, C.H.; Porter, D.L.; et al. Cytokine Release Syndrome After Chimeric Antigen Receptor T Cell Therapy for Acute Lymphoblastic Leukemia. Crit. Care Med. 2017, 45, e124–e131. [Google Scholar] [CrossRef] [PubMed]
  31. Alvi, R.M.; Frigault, M.J.; Fradley, M.G.; Jain, M.D.; Mahmood, S.S.; Awadalla, M.; Lee, D.H.; Zlotoff, D.A.; Zhang, L.; Drobni, Z.D.; et al. Cardiovascular Events Among Adults Treated With Chimeric Antigen Receptor T-Cells (CAR-T). J. Am. Coll. Cardiol. 2019, 74, 3099–3108. [Google Scholar] [CrossRef] [PubMed]
  32. Lefebvre, B.; Kang, Y.; Smith, A.M.; Frey, N.V.; Carver, J.R.; Scherrer-Crosbie, M. Cardiovascular Effects of CAR T Cell Therapy. JACC Cardio Oncol. 2020, 2, 193–203. [Google Scholar] [CrossRef] [PubMed]
  33. Ganatra, S.; Parikh, R.; Neilan, T.G. Cardiotoxicity of Immune Therapy. Cardiol. Clin. 2019, 37, 385–397. [Google Scholar] [CrossRef]
  34. Aldoss, I.; Khaled, S.K.; Budde, E.; Stein, A.S. Cytokine Release Syndrome with the Novel Treatments of Acute Lymphoblastic Leukemia: Pathophysiology, Prevention, and Treatment. Curr. Oncol. Rep. 2019, 21, 4. [Google Scholar] [CrossRef] [PubMed]
  35. Ganatra, S.; Carver, J.R.; Hayek, S.S.; Ky, B.; Leja, M.J.; Lenihan, D.J.; Lenneman, C.; Mousavi, N.; Park, J.H.; Perales, M.A.; et al. Chimeric Antigen Receptor T-Cell Therapy for Cancer and Heart. J. Am. Coll. Cardiol. 2019, 74, 3153–3163. [Google Scholar] [CrossRef] [PubMed]
  36. Jamal, F.A.; Khaled, S.K. The Cardiovascular Complications of Chimeric Antigen Receptor T Cell Therapy. Curr. Hematol. Malign-Rep. 2020, 15, 130–132. [Google Scholar] [CrossRef] [PubMed]
  37. Dal’Bo, N.; Patel, R.; Parikh, R.; Shah, S.P.; Guha, A.; Dani, S.S.; Ganatra, S. Cardiotoxicity of Contemporary Anticancer Immunotherapy. Curr. Treat. Options Cardiovasc. Med. 2020, 22, 1–15. [Google Scholar] [CrossRef] [PubMed]
  38. Asnani, A. Cardiotoxicity of Immunotherapy: Incidence, Diagnosis, and Management. Curr. Oncol. Rep. 2018, 20, 1–7. [Google Scholar] [CrossRef]
  39. Neelapu, S.S.; Tummala, S.; Kebriaei, P.; Wierda, W.; Gutierrez, C.; Locke, F.L.; Komanduri, K.V.; Lin, Y.; Jain, N.; Daver, N.; et al. Chimeric antigen receptor T-cell therapy—Assessment and management of toxicities. Nat. Rev. Clin. Oncol. 2018, 15, 47–62. [Google Scholar] [CrossRef]
  40. Murthy, H.; Iqbal, M.; Chavez, J.C.; Kharfan-Dabaja, M.A. Cytokine Release Syndrome: Current Perspectives. ImmunoTargets Ther. 2019, 8, 43–52. [Google Scholar] [CrossRef] [Green Version]
  41. Riegler, L.L.; Jones, G.P.; Lee, D.W. Current approaches in the grading and management of cytokine release syndrome after chimeric antigen receptor T-cell therapy. Ther. Clin. Risk Manag. 2019, 15, 323–335. [Google Scholar] [CrossRef] [Green Version]
  42. Le, R.Q.; Li, L.; Yuan, W.; Shord, S.S.; Nie, L.; Habtemariam, B.A.; Przepiorka, D.; Farrell, A.T.; Pazdur, R. FDA Approval Summary: Tocilizumab for Treatment of Chimeric Antigen Receptor T Cell-Induced Severe or Life-Threatening Cytokine Release Syndrome. Oncologist 2018, 23, 943–947. [Google Scholar] [CrossRef] [Green Version]
  43. Mahmoudjafari, Z.; Hawks, K.G.; Hsieh, A.A.; Plesca, D.; Gatwood, K.S.; Culos, K.A. American Society for Blood and Marrow Transplantation Pharmacy Special Interest Group Survey on Chimeric Antigen Receptor T Cell Therapy Administrative, Logistic, and Toxicity Management Practices in the United States. Biol. Blood Marrow Transpl. 2019, 25, 26–33. [Google Scholar] [CrossRef] [Green Version]
  44. Lobenwein, D.; Kocher, F.; Dobner, S.; Gollmann-Tepeköylü, C.; Holfeld, J. Cardiotoxic mechanisms of cancer immunotherapy—A systematic review. Int. J. Cardiol. 2021, 323, 179–187. [Google Scholar] [CrossRef]
  45. Annane, D.; Maxime, V.; Ibrahim, F.; Alvarez, J.C.; Abe, E.; Boudou, P. Diagnosis of Adrenal Insufficiency in Severe Sepsis and Septic Shock. Am. J. Respir. Crit. Care Med. 2006, 174, 1319–1326. [Google Scholar] [CrossRef] [Green Version]
  46. Dellinger, R.P.; Levy, M.M.; Carlet, J.M.; Bion, J.; Parker, M.M.; Jaeschke, R.; Reinhart, K.; Angus, D.C.; Brun-Buisson, C.; Beale, R.; et al. Surviving Sepsis Campaign: International guidelines for management of severe sepsis and septic shock: 2008. Crit. Care Med. 2008, 36, 296–327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Bethin, K.E.; Vogt, S.K.; Muglia, L.J. Interleukin-6 is an essential, corticotropin-releasing hormone-independent stimulator of the adrenal axis during immune system activation. Proc. Natl. Acad. Sci. USA 2000, 97, 9317–9322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Buckley, L.F.; Abbate, A. Interleukin-1 blockade in cardiovascular diseases: A clinical update. Eur. Hear. J. 2018, 39, 2063–2069. [Google Scholar] [CrossRef] [Green Version]
  49. 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. 2018, 24, 739–748. [Google Scholar] [CrossRef]
  50. Giavridis, T.; Van Der Stegen, S.J.C.; Eyquem, J.; Hamieh, M.; Piersigilli, A.; Sadelain, M. CAR T cell–induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat. Med. 2018, 24, 731–738. [Google Scholar] [CrossRef] [PubMed]
  51. Jatiani, S.S.; Aleman, A.; Madduri, D.; Chari, A.; Cho, H.J.; Richard, S.; Richter, J.; Brody, J.; Jagannath, S.; Parekh, S. Myeloma CAR-T CRS Management With IL-1R Antagonist Anakinra. Clin. Lymphoma Myeloma Leuk. 2020, 20, 632–636.e1. [Google Scholar] [CrossRef]
  52. Wang, J.; Mou, N.; Yang, Z.; Li, Q.; Jiang, Y.; Meng, J.; Liu, X.; Deng, Q. Efficacy and safety of humanized anti-CD19CAR-T therapy following intensive lymphodepleting chemotherapy for refractory/relapsed B acute lymphoblastic leukaemia. Br. J. Haematol. 2020, 191, 212–222. [Google Scholar] [CrossRef] [Green Version]
  53. Zhang, L.; Wang, S.; Xu, J.; Zhang, R.; Zhu, H.; Wu, Y.; Zhu, L.; Li, J.; Chen, L. Etanercept as a new therapeutic option for cytokine release syndrome following chimeric antigen receptor T cell therapy. Exp. Hematol. Oncol. 2021, 10, 1–4. [Google Scholar] [CrossRef] [PubMed]
  54. Rodríguez-Otero, P.; Prósper, F.; Alfonso, A.; Paiva, B.; Miguel, J.F.S.S. CAR T-Cells in Multiple Myeloma Are Ready for Prime Time. J. Clin. Med. 2020, 9, 3577. [Google Scholar] [CrossRef] [PubMed]
  55. Grupp, S.A.; Kalos, M.; Barrett, D.; Aplenc, R.; Porter, D.L.; Rheingold, S.R.; Teachey, D.T.; Chew, A.; Hauck, B.; Wright, J.F.; et al. Chimeric Antigen Receptor–Modified T Cells for Acute Lymphoid Leukemia. N. Engl. J. Med. 2013, 368, 1509–1518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Summary of CAR-T formulation and administration.
Figure 1. Summary of CAR-T formulation and administration.
Diseases 09 00020 g001
Figure 2. Pre-operative assessment. CAR-T: Chimeric antigen receptor T-cells; EKG: Electrocardiogram; TTE: Transthoracic echocardiogram; CAD: Coronary artery disease.
Figure 2. Pre-operative assessment. CAR-T: Chimeric antigen receptor T-cells; EKG: Electrocardiogram; TTE: Transthoracic echocardiogram; CAD: Coronary artery disease.
Diseases 09 00020 g002
Figure 3. Management consideration in patients that develop CRS that may be at risk for cardiotoxicity. CRS: Cytokine release syndrome. TTE: Transthoracic echocardiogram; EKG: Electrocardiogram; EF: Ejection fraction; proBNP: Brain natriuretic peptide.
Figure 3. Management consideration in patients that develop CRS that may be at risk for cardiotoxicity. CRS: Cytokine release syndrome. TTE: Transthoracic echocardiogram; EKG: Electrocardiogram; EF: Ejection fraction; proBNP: Brain natriuretic peptide.
Diseases 09 00020 g003
Table 1. Grading Criterias for CRS in the pivotal and included retrospective studies.
Table 1. Grading Criterias for CRS in the pivotal and included retrospective studies.
Penn Criteria [12]
Grade 1Grade 2Grade 3Grade 4
Mild reaction: Treated with supportive care, such as antipyretics, antiemeticsModerate reaction: Some signs of organ dysfunction (grade 2 creatinine or grade 3 LFTs) related to CRS and not attributable to any other condition. Hospitalization for management of CRS-related symptoms, including neutropenic fever and need for i.v. therapies (not including fluid resuscitation for hypotension)More severe reaction: Hospitalization required for management of symptoms related to organ dysfunction, including grade 4 LFTs or grade 3 creatinine, related to CRS and not attributable to any other condition. Hypotension treated with multiple fluid boluses or low-dose vasopressors. Coagulopathy requiring fresh frozen plasma, cryoprecipitate, or fibrinogen concentrate. Hypoxia requiring supplemental oxygen (nasal cannula oxygen, high-flow oxygen, CPAP, or BiPAP)Life-threatening complications such as hypotension requiring high-dose vasopressors. Hypoxia requiring mechanical ventilation
Lee Criteria [13]
Grade 1Grade 2Grade 3Grade 4
Symptoms are not life-threatening and require symptomatic treatment only (fever, nausea, fatigue, headache, myalgias, malaise)Symptoms require and respond to moderate intervention:
Oxygen requirement < 40% FiO2, OR hypotension responsive to i.v. fluids or low dose of one vasopressor, OR grade 2 organ toxicity *
Symptoms require and respond to aggressive intervention:
Oxygen requirement ≥ 40% FiO2, OR Hypotension requiring high-dose or multiple vasopressors, OR grade 3 organ toxicity, or grade 4 transaminitis
Life-threatening symptoms:
Requirement for ventilator support, OR grade 4 organ toxicity (excluding transaminitis)
ASTCT Consensus Criteria [14]
Grade 1Grade 2Grade 3Grade 4
Temperature ≥ 38 °C, no hypotension, no hypoxiaTemperature ≥ 38 °C, with hypotension not requiring vasopressors, and/or hypoxia requiring low flow nasal cannulaTemperature ≥ 38 °C, with hypotension requiring vasopressors with or without vasopressin, and/or hypoxia requiring high-flow nasal cannula, facemask, nonrebreather mask, or venturi maskTemperature ≥ 38 °C, with hypotension requiring multiple vasopressors (excluding vasopressin), and/or hypoxia requiring positive pressure (CPAP, BiPAP, intubation, and mechanical ventilation)
* Cardiac (tachycardia, arrhythmias, heart block, or decrease in ejection fraction), respiratory (tachypnea, pleural effusion, or pulmonary edema), renal (acute kidney injury, increase in serume creatinine level, or decrease in urine output), gastrointestinal (nausea, vomiting, or diarrhea), hepatic (increase in serum alanine aminotransferase, aspartate aminotransferase, or bilirubin level), coagulopathy (disseminated intravascular coagulation), or dermatologic (rash).
Table 2. Summary of reported cardiovascular events associated with CD19-CAR-T.
Table 2. Summary of reported cardiovascular events associated with CD19-CAR-T.
Reported Cardiotoxic Events with FDA Approved CD19 CAR-T
Tachycardia
Hypotension
Fluid refractory hypotension
Pulmonary Edema
Depressed left ventricular function
Cardiac failure
Cardiac failure requiring inotropic support
Elevated troponin
Arrhythmia
ST-segment changes
Cardiac arrest
Table 3. Cardiac exclusion criteria for pivotal trials of CD19-CAR-T. ECG: Electrocardiogram.
Table 3. Cardiac exclusion criteria for pivotal trials of CD19-CAR-T. ECG: Electrocardiogram.
CD19-CAR-T InfusionTisagenlecleucel TisagenlecleucelAxicabtagene CiloleucelBrexucabtagene Autoleucel
Trial ELIANA [8]JULIET [7]ZUMA-1 [9]ZUMA-2 [10]
Pertinent cardiovascular trial exclusion criteria-Left Ventricular systolic function ≤ 28% confirmed by echocardiogram
-Left ventricular ejection fraction ≤ 45% confirmed by echocardiogram or multigated acquisition images within 7 days of screening
-Unstable Angina or MI within 6 months of planned infusion
-Uncontrolled arrhythmia
-EF < 50% determined by transthoracic echocardiogram
-Evidence of pericardial effusion
-Presence of clinically significant ECG findings
-Cardiac ejection fraction < 50%
-Evidence of pericardial effusion
-Clinically significant electrocardiogram findings
-Myocardial infarction, cardiac angioplasty or stenting, unstable angina, active arrhythmias, or other clinically significant cardiac disease within 12 months of enrollment
-Cardiac atrial or cardiac ventricular lymphoma involvement
Table 4. Summary of outcomes and reported cardiotoxic events in the pivotal phase 2 trials leading to FDA approval of Tisgenlecleucel, Axicabtagene Ciloleucel, and Brexucabtagene Ciloleucel.
Table 4. Summary of outcomes and reported cardiotoxic events in the pivotal phase 2 trials leading to FDA approval of Tisgenlecleucel, Axicabtagene Ciloleucel, and Brexucabtagene Ciloleucel.
FDA Approved CD19-CAR-TTisagenlecleucelTisagenlecleucel Axicabtagene CiloleucelBrexucabtagene Autoleucel
Trial JULIET [7]ELIANA [8]ZUMA-1 [9]ZUMA-2 [10]
DiseaseAdult LBCLPediatric B-ALLAdult LBCLAdult MCL
Study Phase21–222
Patients Studied in Efficacy Analysis937510168
Objective Response Rate50%83%82%93%
Complete Response40%60%54%67%
12 month RFS/PFS65%59%44%61%
12 month OS49% (estimated)76%59%83%
Patients Studied in Safety Analysis1117510168
Percent with any Grade AE100%100%100%100%
CRS64 (58%)58 (77%)94 (93%)61 (91%)
CRS Grading SystemPenn Criteria [12]Penn Criteria [12]Lee Criteria [13]Lee Criteria [13]
Tocilizumab Use16 (14%)36 (48%)49 (48.5%)42 (61.8%)
Hypotension29 (26%)22(29%)60 (59%)35 (51%)
Hypotension requiring inotropic support or shock8 (9%)13 (17%)14 (14%)15 (22%)
Pulmonary EdemaNR5 (6.7%)NRNR
Left Ventricular DysfunctionNR3 (4.0%)NRNR
Cardiac ArrestNR3(4.0%)NRNR
Cardiac FailureNR2 (2.7%)NRNR
Tachycardia12 (11%)3 (4.0%)39 (39%)21 (31%)
B-ALL: B-cell Acute Lymphoblastic Leukemia; RFS: Relapse Free Survival; PFS: Progression Free Survival; OS: Overall Survival; AE: Adverse Events; CRS: Cytokine Release syndrome; LBCL: Large B-cell Lymphoma; MCL: Mantle Cell Lymphoma.
Table 5. Summary of cardiotoxicity in retrospective pediatric assessments.
Table 5. Summary of cardiotoxicity in retrospective pediatric assessments.
CD19-CAR-T Cardiovascular Events Shalabi et al. (2020) [28]Burstein et al. (2018) [29]Fitzgerald et al. (2017) [30]
Patient PopulationPediatric (n = 52)Pediatric (n = 98)Pediatric (n = 39)
Treatment Indication
B-ALL50 (96.1%)90 (97%)39 (100%)
NHL2 (3.9%)1 (1%)0
Multiple Myeloma000
T-ALL01 (1%)0
PML01 (1%)0
CRS Grading SystemPenn Criteria [12]
ASTCT Consensus Criteria [14]
Penn Criteria [12]Penn Criteria [12]
Cardiotoxic Events
Pre-existing Cardiomyopathy/Structural Disease/Arrhythmia6 (11.5%)10 (11%)/1(5%)NR
Hypotension Requiring Inotropic Support9 (24.3%)24 (24%)13 (33%)
TroponemiaNRNRNR
Ventricular Systolic Dysfunction6 (11.5%)10 (10%)1 (2%)
Tachycardia36 (69.2%)NRNR *
ArrhythmiaNRNRNR
ST segment changesNR6 (6%)NR
Cardiac Arrest/ Cardiac Death1 (2.7%)0NR
Required Tocilizumab14 (37.8%)21 (21%)13 (33%)
* Number with tachycardia not reported. B-ALL: B-cell Acute Lymphoblastic Leukemia; NHL: non-Hodgkin Lymphoma; T-ALL: T-cell Acute Lymphoblastic Leukemia; PML: Primary Mediastinal Large B-cell Lymphoma.
Table 6. Summary of cardiotoxicity in retrospective adult assessments.
Table 6. Summary of cardiotoxicity in retrospective adult assessments.
CD19-CAR-T Cardiovascular Events Ganatra et al. (2020) [18]Alvi et al. (2019) [31]Lefebvre et al. (2020) [32]
Patient PopulationAdults (n = 187)Adult (n = 137)Adult (n = 145)
Treatment Indication
B-ALL1 (0.5%)036 (25%)
NHL185 (98.7%)119 (88%)43 (30%)
Multiple Myeloma011 (8%)0
T-ALL000
PML1 (0.5%)00
CLL0066 (46%)
CRS Grading SystemLee Criteria [13]Lee Criteria [13]ASTCT consensus Criteria [14]
Number with Cardiotoxic event12 (6.4%)17 (12%)31 (21.3%)
Pre-existing Cardiomyopathy/Structural Disease/Arrhythmia1 (0.5%)/4 (2.1%)/3 (1.6%)5 (3.6%)/10 (7.3%)/18 (13%)1 (0.7%)/5 (3.4%)/5 (3.4%)
Hypotension/shock Requiring Inotropic Support5 (2.6%)6 (4%)33 (22.7%)
TroponemiaNR29 (21%)NR
CHF/Ventricular Systolic Dysfunction12 (6.4%)8 (6%)21 (14.5%)
Sinus TachycardiaNR6 (4.4%)NR
Arrhythmia 5 (3.6%)13 (8.9%)
ST segment changesNRNRNR
Cardiac Arrest/ Cardiac Death3 (1.6%)6 (4.4%)2 (1.4%)
Required Tocilizumab12 (6.4%)56 (40.9%)15 (10.3%)
B-ALL: B-cell Acute Lymphoblastic Leukemia; NHL: non-Hodgkin Lymphoma; T-ALL: T-cell Acute Lymphoblastic Leukemia; PML: Primary Mediastinal Large B-cell Lymphoma. CLL: Chronic Lymphocytic Leukemia; CHF: Congestive Heart Failure.
Table 7. Baseline factors that may increase the risk of Cardiotoxicity.
Table 7. Baseline factors that may increase the risk of Cardiotoxicity.
Predictive Risk Factors for CRS [25,27,34,35]Risk Factors for Cardiotoxicity in Pediatric Patients [28,29]Risk Factors for Cardiotoxicity in Adult Patients [18,31,32,35]
High disease burdenPre-Treatment Blasts >25% on bone marrow biopsyConcomitant CRS (grade 3 or 4 CRS)
High CAR-T doseLower Pre-CAR-T Treatment baseline EFTroponin elevation
High intensity lymphodepleting regimenPre-existing diastolic dysfunctionOlder Age
Pre-existing endothelial activation Higher Baseline Creatinine
Severe thrombocytopenia Aspirin, statin, insulin, beta blocker, RAA medication use
Addition of fludarabine to cyclophosphamide during lymphodepletion Hyperlipidemia
Higher peak of C reactive protein CAD
Older patient age Aortic Stenosis
EF: Ejection Fraction; CRS: Cytokine Release Syndrome; CAD: Coronary Artery Disease; RAA: Renin-angiotensin-aldosterone.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Burns, E.A.; Gentille, C.; Trachtenberg, B.; Pingali, S.R.; Anand, K. Cardiotoxicity Associated with Anti-CD19 Chimeric Antigen Receptor T-Cell (CAR-T) Therapy: Recognition, Risk Factors, and Management. Diseases 2021, 9, 20. https://0-doi-org.brum.beds.ac.uk/10.3390/diseases9010020

AMA Style

Burns EA, Gentille C, Trachtenberg B, Pingali SR, Anand K. Cardiotoxicity Associated with Anti-CD19 Chimeric Antigen Receptor T-Cell (CAR-T) Therapy: Recognition, Risk Factors, and Management. Diseases. 2021; 9(1):20. https://0-doi-org.brum.beds.ac.uk/10.3390/diseases9010020

Chicago/Turabian Style

Burns, Ethan A., Cesar Gentille, Barry Trachtenberg, Sai Ravi Pingali, and Kartik Anand. 2021. "Cardiotoxicity Associated with Anti-CD19 Chimeric Antigen Receptor T-Cell (CAR-T) Therapy: Recognition, Risk Factors, and Management" Diseases 9, no. 1: 20. https://0-doi-org.brum.beds.ac.uk/10.3390/diseases9010020

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop