Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Biology of Blood and Marrow Transplantation, Elsevier BV, Vol. 21, No. 2 ( 2015-02), p. S309-
    Type of Medium: Online Resource
    ISSN: 1083-8791
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2015
    detail.hit.zdb_id: 3056525-X
    detail.hit.zdb_id: 2057605-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Biology of Blood and Marrow Transplantation, Elsevier BV, Vol. 25, No. 3 ( 2019-03), p. S108-S109
    Type of Medium: Online Resource
    ISSN: 1083-8791
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2019
    detail.hit.zdb_id: 3056525-X
    detail.hit.zdb_id: 2057605-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Biology of Blood and Marrow Transplantation, Elsevier BV, Vol. 24, No. 3 ( 2018-03), p. S75-S76
    Type of Medium: Online Resource
    ISSN: 1083-8791
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2018
    detail.hit.zdb_id: 3056525-X
    detail.hit.zdb_id: 2057605-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 39, No. 15 ( 2021-05-20), p. 1650-1659
    Abstract: CD19 chimeric antigen receptor (CD19-CAR) T cells induce high response rates in children and young adults (CAYAs) with B-cell acute lymphoblastic leukemia (B-ALL), but relapse rates are high. The role for allogeneic hematopoietic stem-cell transplant (alloHSCT) following CD19-CAR T-cell therapy to improve long-term outcomes in CAYAs has not been examined. METHODS We conducted a phase I trial of autologous CD19.28ζ-CAR T cells in CAYAs with relapsed or refractory B-ALL. Response and long-term clinical outcomes were assessed in relation to disease and treatment variables. RESULTS Fifty CAYAs with B-ALL were treated (median age, 13.5 years; range, 4.3-30.4). Thirty-one (62.0%) patients achieved a complete remission (CR), 28 (90.3%) of whom were minimal residual disease−negative by flow cytometry. Utilization of fludarabine/cyclophosphamide–based lymphodepletion was associated with improved CR rates (29/42, 69%) compared with non–fludarabine/cyclophosphamide–based lymphodepletion (2/8, 25%; P = .041). With median follow-up of 4.8 years, median overall survival was 10.5 months (95% CI, 6.3 to 29.2 months). Twenty-one of 28 (75.0%) patients achieving a minimal residual disease−negative CR proceeded to alloHSCT. For those proceeding to alloHSCT, median overall survival was 70.2 months (95% CI, 10.4 months to not estimable). The cumulative incidence of relapse after alloHSCT was 9.5% (95% CI, 1.5 to 26.8) at 24 months; 5-year EFS following alloHSCT was 61.9% (95% CI, 38.1 to 78.8). CONCLUSION We provide the longest follow-up in CAYAs with B-ALL after CD19-CAR T-cell therapy reported to date and demonstrate that sequential therapy with CD19.28ζ-CAR T cells followed by alloHSCT can mediate durable disease control in a sizable fraction of CAYAs with relapsed or refractory B-ALL (ClinicalTrials.gov identifier: NCT01593696 ).
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2021
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Pediatric Blood & Cancer, Wiley, Vol. 63, No. 6 ( 2016-06), p. 997-1005
    Abstract: Vincristine sulfate liposome injection (VSLI; Marqibo®) is an encapsulated preparation of standard vincristine in sphingomyelin/cholesterol liposomes. Clinical trials in adults have demonstrated safety, tolerability, and activity, leading to Food and Drug Administration (FDA) approval for adults with relapsed acute lymphoblastic leukemia (ALL). Pediatric experience with VSLI is limited. Procedure This single center, phase I dose escalation study examined the safety, toxicity, maximum tolerated dose, and pharmacokinetics of VSLI administered weekly to pediatric patients age 〈 21 years with relapsed or chemotherapy‐refractory solid tumors or leukemia. Results Twenty‐one subjects were treated in total. Median age was 13.3 years (range 2–19). Fourteen subjects completed one 28‐day cycle of therapy and five subjects completed more than one cycle. No subject experienced dose‐limiting toxicity (DLT) at the first dose level (1.75 mg/m 2 /dose, dose range: 2–3.7 mg). At the second dose level (2.25 mg/m 2 /dose, dose range: 1.3–4.5 mg), one subject had transient dose‐limiting grade 4 transaminase elevation, and this dose level was expanded with no additional DLT observed. The second dose level then opened to an expansion phase to evaluate activity in ALL. Clinical activity included minimal residual disease negative complete remission in one subject with ALL and stable disease in nine subjects. Clearance of total vincristine was found to be approximately 100‐fold lower in comparison to published data using standard vincristine. Conclusions Children tolerate 2.25 mg/m 2 /dose of weekly VSLI (the adult FDA‐approved dose) with evidence for clinical activity without dose‐limiting neurotoxicity. Future plans include studying VSLI as substitution for standard vincristine with combination chemotherapy in children with ALL.
    Type of Medium: Online Resource
    ISSN: 1545-5009 , 1545-5017
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2016
    detail.hit.zdb_id: 2130978-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Leukemia, Springer Science and Business Media LLC, Vol. 34, No. 11 ( 2020-11), p. 3064-3069
    Type of Medium: Online Resource
    ISSN: 0887-6924 , 1476-5551
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2008023-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 2609-2609
    Abstract: Abstract 2609 Children with relapsed or chemotherapy-refractory ALL have a poor prognosis despite the use of aggressive therapies such as HSCT. Chimeric antigen receptor modified T cells targeting the B-cell antigen CD19 have been reported to be effective in adults with B-cell lymphomas and chronic lymphocytic leukemia. We conducted pre-clinical studies with a CD19-CAR consisting of a CD19-specific scFv and the CD28 and CD3z signaling domains. These cells generated significant levels of IFNg, TNFa, and IL-2 in response to ALL blasts and rapidly eradicated human ALL in murine xenografts. We developed a Phase I clinical trial of CD19-CAR modified autologous T cells for children with CD19+ hematologic malignancies. HSCT-na•ve and post-transplant patients are eligible, and cells are collected directly from patients in both cases. CD19-CAR T cells are manufactured in a semi-closed system over an 11-day period. We report results with the first patient, a 13-year old with chemotherapy-refractory ALL that had relapsed after 2 prior matched related donor HSCTs. Peripheral blood (PB) mononuclear cells were collected from the patient on Day -11 by apheresis. T cells were positively selected and activated by incubation with anti-CD3/anti-CD28 paramagnetic beads in IL-2 for 48 hours then transduced with the CD19-CAR gene via retroviral supernatant for an additional 48 hours. Beads were removed and the expanding CD19-CAR T cells were maintained in culture with IL-2 until harvested for infusion on Day 0. A 59-fold expansion of CAR T cells with 65% transduction efficiency was achieved. The patient was pre-treated with fludarabine (25 mg/m2/day on Days -4, -3, -2) and cyclophosphamide (900 mg/m2 on Day -2) prior to the infusion of 1×106 CAR-transduced T cells/kg. The patient developed signs and symptoms of cytokine release syndrome (CRS) on Day +5 with full resolution by Day +11. Manifestations included fever (maximum 41°C), rigors (Grade 1), and hypotension (Grade 2), the latter of which was responsive to two IV fluid boluses. The patient also developed an erythematous rash (Grade 1) of the extremities from Days +7 to +12 and bilateral scrotal swelling and pain (Grade 1) from Days +8 to +10, which was associated with increased testicular blood flow by ultrasound. Cytokine analysis revealed high levels of IL-6 (53.1 pg/ml; normal 〈 5), GM-CSF (59.4 pg/ml; normal 〈 7.8) and IFNg (44.7 pg/ml; normal 〈 15.6). As IL-6 induces release of C-reactive protein (CRP) from the liver, we monitored CRP levels, which mirrored the time course of CRS (Figure). Broad-spectrum antibiotics were administered for neutropenic fever, although all cultures were negative. No other therapeutic interventions were provided aside from routine supportive care. Figure. Maximum temperature in each 24-hour period (Tmax, red solid line) and CRP (blue solid line) correlate with cytokine release syndrome. Dotted lines indicate upper limits of normal. PB flow cytometry performed prior to cell infusion demonstrated 0.04% blasts and 1% normal B-cells. Repeat analysis after infusion revealed clearance of blasts, gradually decreasing B cells, and a maximum of 0.07% CD19CAR T cells (Days +3, +6, and +27). Re-staging evaluation on Day +27 revealed achievement of a complete remission with bone marrow (BM) blasts decreasing from 30% pre to 3% (flow cytometry 5% to 0.6%) and cerebrospinal fluid (CSF) blasts decreasing from 1.5% to 0%. 0.4% of BM T cells expressed the CD19CAR. (Table) Figure. Maximum temperature in each 24-hour period (Tmax, red solid line) and CRP (blue solid line) correlate with cytokine release syndrome. Dotted lines indicate upper limits of normal. PB flow cytometry performed prior to cell infusion demonstrated 0.04% blasts and 1% normal B-cells. Repeat analysis after infusion revealed clearance of blasts, gradually decreasing B cells, and a maximum of 0.07% CD19CAR T cells (Days +3, +6, and +27). Re-staging evaluation on Day +27 revealed achievement of a complete remission with bone marrow (BM) blasts decreasing from 30% pre to 3% (flow cytometry 5% to 0.6%) and cerebrospinal fluid (CSF) blasts decreasing from 1.5% to 0%. 0.4% of BM T cells expressed the CD19CAR. (Table) Day % BM Blasts (Flow) BM CAR % of T cells % PB Blasts PB CAR % of T cells PB B Cells % % CSF Blasts % CSF CAR Pre 30 (5) 0 0.04 0 1 1.5 0 +3 0 0.07 0.9 +6 0 0.07 0.2 +13 0 0 0.1 +27 3 (0.6) 0.4 0.01 0.07 0.06 0 0 Conclusions: A complete remission was successfully induced in a child with chemotherapy-refractory ALL and post-transplant relapse using a single infusion of autologous-collected, donor-derived T cells modified with a CD19-CAR. Treatment was well tolerated and was only associated with mild CRS that was characterized by high levels of IL-6, INFg, and GM-CSF but not IL-1b or TNFa. CRP levels can be used as a readily available biomarker of IL-6-associated CRS. To our knowledge, this is the first CAR-based therapy to be utilized in the post-allogeneic setting using donor-derived T cells collected directly from a pediatric patient. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 684-684
    Abstract: CD19 chimeric antigen receptor (CAR) T cells have shown significant promise in multiple early phase trials including our own (Lancet 385:517-28). We manufacture CAR T cells containing CD28 and CD3z domains in 7 days using a retroviral platform. Several challenges remain to its widespread use: 1) reduction in the incidence of grade 4 cytokine release syndrome (CRS) and 2) incorporation with standard salvage regimens. Here, we update our experience with 39 patients. In the first 21 patients we defined the maximally tolerated dose as 1x106 CAR T cells/kg, grade 4 CRS occurred in 16%, and noted that severity of CRS correlated with disease burden. We stratified the current cohort (n=18) by disease burden. Subjects 1-21 and subsequent patients with low burden disease (Arm 1: isolated CNS disease or 〈 25% marrow blasts) received a low dose preparative regimen of fludarabine (25 mg/m2/day Days-4 to -2) and cyclophosphamide (900 mg/m2 Day-2). Those with high burden disease (Arm 2: ³25% marrow blasts, circulating blasts or lymphomatous disease) received a high dose regimen to reduce tumor burden prior to cell infusion in an attempt to decrease severity of CRS. Arm 2 regimens were individualized based on prior therapies and risk from comorbidities. FLAG (n=6), ifosfamide/etoposide per AALL0031 (IE; n=2) and high dose fludarabine (30 mg/m2/day Days -6 to -3) with cyclophosphamide (1200 mg/m2/day Days -4 and -3) (HD flu/cy; n=3) were used. All products in the second cohort met cell dose though contaminating monocytes tended to inhibit maximal growth and transduction (see companion abstract by Stroncek). All patients received 1x106 CAR T cells/kg. Using grading criteria and an algorithm for early intervention to prevent grade 4 CRS (Blood 124:188-95) no grade 3 and only 1 grade 4 (5.6%) CRS occurred. Having significant comorbidities, Pt 34 was electively intubated for airway protection, did not require vasopressors, and rapidly recovered after tocilizumab and steroids. A brief seizure occurred, though he had a history of seizures. None others in the current cohort had neurotoxicity. Using intent to treat analysis, the complete response (CR) rate was 59% overall and 61% in ALL. 13/16 (81%) low burden and 10/22 (46%) high burden ALL patients had a CR across both cohorts. Low burden patients treated on either cohort had similar CR rate of 8/10 (80%) and 5/6 (83%). Although not statistically significant and underpowered, 7/11 (64%) high burden patients treated with low dose flu/cy had a CR while 3/11 (27%) had a CR with high dose regimens. Specifically, 3/6 (50%) receiving FLAG achieved MRD-CR while none receiving IE or HD flu/cy responded. 8/8 with primary refractory ALL had MRD-CR regardless of disease burden or preparative regimen raising the prospect that T cell fitness in these patients was superior to others. Of the 20 patients achieving an MRD-CR, the median leukemia free survival (LFS) is 17.7 months with 45.5% probability of LFS beginning at 18 months. Only 3 did not have a subsequent hematopoietic stem cell transplant as their referring oncologist determined the risk of such was unacceptable. Two relapsed with CD19-leukemia at 3 and 5 months, while 1 remains in CR with detectable CAR T cells at 5 months. Reliance on multiple infusions of cells is problematic as 0/5 CD19+ patients receiving a second dose responded. Preclinical models have demonstrated that T cell exhaustion has a role in limiting the efficacy of CAR T cells. We evaluated CAR products and the T cells used to generate them for phenotypic markers of exhaustion and will present data evaluating the relationship between these and response. Our results demonstrate that CD19 CAR T cell therapy is safe and effective with aggressive supportive care and use of an early intervention algorithm to prevent severe CRS and provides a potential for cure in primary refractory ALL. Table. Patient Characteristics, Response, and Toxicity Pt Age/ Sex/Risk # Relapses Arm/Prep Regimen(if Arm 2) Marrow Blasts Response CRS Grade Pre-Therapy Post CAR 22 17M 3 1 20 0 MRD- 2 23 13M 2 2 IE 99 98 SD 0 24 12M MLL 2 1 8.5 3 CR 1 25 25F 1 2 FLAG 95 0 MRD- 2 26 4M DS 2 2 IE (60%) 89 NA PD 0 27 8F 2 2 FLAG 77 69 SD 0 28 4M 2 2 FLAG (60%) 99 99 PD 0 29 12M PR 1 0.15 0 MRD- 1 30 15M Ph+ CNS2 3 1 0.08 0 MRD- 1 31 22M 3 2 FLAG 97 99 SD 0 32 15M CNS2 3 2 FLAG 0.04 + Lymphoma 0 MRD- 2 33 6M PR 1 0.15 0 MRD- 0 34 14M DS 3 2 Arm 1 Flu/Cy 90 0 MRD- 4 35 25M 2 2 HD Flu/Cy 30 87 PD 2 36 6M 2 1 1.5 91 PD 0 37 4F MLL 1 2 HD Flu/Cy 90 99 SD 0 38 7M 1 2 HD Flu/Cy 99 99 SD 1 Disclosures Off Label Use: Off-label use of tocilizumab will be discussed in managing cytokine release syndrome.. Rosenberg:Kite Pharma: Other: CRADA between Surgery Branch-NCI and Kite Pharma. Mackall:Juno: Patents & Royalties: CD22-CAR.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 218-218
    Abstract: Relapsed pre-B acute lymphoblastic leukemia (ALL) portends a poor prognosis even with hematopoietic stem cell transplantation (HSCT). CD19 chimeric antigen receptor (CAR) T cells have shown promise in early studies although morbidity related tohigh gradecytokine release syndrome (CRS) and/or neurotoxicity could limit its wide applicability in patients with high disease burden. The lympho depleting chemotherapy regimen may affect both toxicity and response and has not been well studied. Relapse rates among complete responders to CD19 CAR therapy occur in nearly half of patients in the first year. We report outcomes from our completed clinical trial of 53 children and young adults with relapsed/refractory ALL (n=51) or lymphoma (n=2) with a median follow up (mF/U) of 18.7 months. The first 21 patients received a low dose fludarabine (25 mg/m2/day Days -4 to -2) and cyclophosphamide (900 mg/m2 Day -2) preparative regimen (LDflu/cy) and results are reported in Lancet 385:517-28. The regimen for the subsequent 32 patients, who all received 1x106 CAR+ T cells/kg, was stratified based on disease burden. Subjects with low burden ALL (lowALL; 〈 25% marrow blasts) received LDflu/cy while those with high burden disease (highALL; 〉 25% marrow blasts or lymphomatous disease) received an alternative regimen [FLAG (n=6), ifosfamide/etoposide per AALL0031 (n=2) or fludarabine (30mg/m2/day Days -6 to -3) and cyclophosphamide (1200 mg/m2/day Days -4 and -3) (HDflu/cy; n=8)] in an attempt to mitigate severe CRS risk and improve response. Four highALL subjects received LDflu/cy due to comorbidities including Trisomy 21. CRS was graded and anti-cytokine therapy was instituted as per Blood 124:188-95. Date for data cutoff was July 31, 2016. Of the 53 subjects 11 had primary refractory ALL, 5Ph+, 3 with Trisomy 21, 4 with CNS2 and 2 with CNS3 ALL including one with extensive leptomeningeal and parenchymal involvement. Cells were manufactured in 7-11 days and none underwent a test expansion. One patient was not infused due to rapidly progressive fungal pneumonia but was accounted for in all analyses. Of 51 ALL patients, 31 (60.8%) achieved a complete response (CR) with 28/31 (90%) of responders negative for minimal residual disease (MRD-). All 6 subjects with CNS ALL were rendered into CNS1 status with resolution of leptomeningeal enhancement, where appropriate, and CAR cells in CSF. The median leukemia free survival (mLFS) of MRD- CR responders is 18 months with a 49.5% probability of LFS beginning at 18 months (mF/U 22.6 months). Grade 3 (n=5) and 4 (n=2) CRS combined for a severe CRS incidence of 13.5%. Three grade 3 neurotoxicities(1 each: dysphasia, delirium, headache) and 2 seizures (one grade 1, one grade 2) occurred. There were no grade 4 neurotoxicities, even in the subject with extensive CNS disease. Subjects with low ALL had a significantly higher CR rate (18/21; 85.7%) than those with high ALL (13/32; 40.6%) (p=0.0011) and use of a flu/cy regimen correlated with higher response (29/44; 65.9% vs 2/8; 25%; p=0.0301). Overall survival in all subjects receiving a flu/cy regimen was 13.3 months with a 34.7% probability of survival beginning at 38 months (mF/U 18.7 months), which is significantly longer than those who did not receive a flu/cy regimen (5.5 months, no survivors beyond 11 months). The hazard ratio (HR) of not receiving a flu/cy regimen was 6.35 (1.906-21.14; p=0.0026). mLFS of subjects with MRD- CR who received a flu/cy regimen was not reached with a 53.3% probability of LFS beginning at 18 months (mF/U 22.6 months). Of the 28 subjects achieving MRD- CR, 21 had a subsequent HSCT with a median time to HSCT of 54 days from CAR infusion. 8/28 (28.6%) relapsed with CD19+ (n=2), CD19-/dim (n=5), CD19 unknown (n=1) blasts. Relapse was significantly more common in subjects who did not have a HSCT after CAR therapy (6/7; 85.7%) compared to those who did (2/21; 9.5%) (p=0.0001). Even accounting for transplant related mortality, them LFS in the HSCT group was not reached with a 62% probability of LFS beginning at 18 months. This is significantly longer than them LFS of 4.9 months in MRD- CR subjects who did not proceed to HSCT (p=0.0006) with a HR of 16.9 (3.37-85.1) of not having a subsequent HSCT. In all, CD19 CAR T cell therapy was effective and safe with a low incidence of severe CRS and neurotoxicity. In this nonrandomized series, the rate of durable remission was higher when a flu/cy preparative regimen was used and consolidation HSCT was employed. Figure 1 Figure 1. Figure 2 Figure 2. Disclosures Lee: Juno: Honoraria. Kochenderfer:bluebird bio: Patents & Royalties, Research Funding; Kite Pharma: Patents & Royalties, Research Funding. Rosenberg:Kite pharma: Research Funding. Mackall:NCI: Patents & Royalties: B7H3 CAR.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 8, No. 2 ( 2020-09), p. e001159-
    Abstract: Chimeric antigen receptor (CAR) T-cell-associated cytokine release syndrome (CRS) may present with tachycardia, hemodynamic instability and reduced cardiac function. Pediatric CAR studies examining cardiac toxicity are limited. Methods We report on cardiac toxicity observed in children and young adults with hematologic malignancies enrolled in a CD19-28ζ CAR T-cell phase I trial ( NCT01593696 ). All patients had a formal baseline echocardiogram. Real-time studies included echocardiograms on intensive care unit (ICU) transfer, and serial troponin and pro-B-type natriuretic peptide (pro-BNP) in the select patients. Results From July 2012 to March 2016, 52 patients, with a median age of 13.4 years (range 4.2–30.3) were treated. CRS developed in 37/52 (71%), which was grade 3–4 CRS in nine patients (17%). The median prior anthracycline exposure was 205 mg/m 2 (range 70–620 mg/m 2 ) in doxorubicin equivalents. The median baseline left ventricle ejection fraction (LVEF) and baseline LV global longitudinal strain (GLS) were 60% (range 50%–70%) and 16.8% (range 14.1%–23.5%, n=37) respectively. The majority, 78% (29/37), of patients had a reduced GLS 〈 19% at baseline, and 6% (3/52) of patients had baseline LVEF 〈 53%. ICU transfers occurred in 21 patients, with nine requiring vasoactive hemodynamic support and three necessitating 〉 1 vasopressor. Six (12%) patients developed cardiac dysfunction (defined by 〉 10% absolute decrease in LVEF or new-onset grade 2 or higher LV dysfunction, per CTCAE v4), among whom 4 had grade 3–4 CRS. Troponin elevations were seen in 4 of 13 patients, all of whom had low LVEF. Pro-BNP was elevated from baseline in 6/7 patients at the onset of CRS, with higher levels correlating with more severe CRS. Cardiac dysfunction fully resolved in all but two patients by day 28 post-CAR. Conclusion Cardiac toxicity related to CD19-28ζ CAR T-cell-associated CRS was generally reversible by day 28 postinfusion. Implementation of more frequent monitoring with formal echocardiograms incorporating systemic analysis of changes in GLS, and cardiac biomarkers (troponin and proBNP) may help to earlier identify those patients at highest risk of severe cardiac systolic dysfunction, facilitating earlier interventions for CRS to potentially mitigate acute cardiac toxicity.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2020
    detail.hit.zdb_id: 2719863-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages