Study Evaluating the Safety, Tolerability, and Efficacy of FLT3 CAR-T AMG 553 in FLT3-positive Relapsed/Refractory AML

Author(s):  
Keyword(s):  
Car T ◽  
Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 811-811
Author(s):  
Lihua Budde ◽  
Joo Y Song ◽  
Young Kim ◽  
Suzette Blanchard ◽  
Jamie Wagner ◽  
...  

Abstract The current treatment of relapsed or refractory AML is associated with low rates of complete response (CR) and considerable complications. As a result, only a minority of patients (pts) proceed to allogeneic hematopoietic stem cell transplantation (alloHSCT) with curative intent. Furthermore, outcomes for AML pts with disease relapse after alloHSCT are dismal. Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare and incurable blood cancer with a median survival of <18 months and no standard of care. Thus, there are clear unmet therapeutic needs in both these conditions. CD123 is overexpressed on AML blasts and leukemic stem cell (LSC)-enriched cell subpopulations compared to normal hematopoietic stem cells and myeloid progenitors. High levels of CD123 expression is also one of the diagnostic hallmarks for BPDCN. All these features make CD123 an attractive target for T cell based adoptive immunotherapy. We have previously demonstrated preclinically the anti-AML activity of CD123-chimeric antigen receptor (CAR) T cell therapy (Mardiros, Blood 2013). The CD123CAR contains an anti-CD123 single-chain variable fragment, an optimized IgG4 CH2CH3 linker, a CD28 co-stimulatory domain, and a CD3 zeta signaling domain and is used to engineer T cells for patients enrolled on a single center, first-in-human phase I dose escalation clinical trial open at our Institution (NCT02159495). The primary objective is to test the safety and activity of escalating doses of CD123CAR T cells for patients with relapsed or refractory AML (cohort 1) and BPDCN (cohort 2). Donor-derived or autologous T cells from leukapheresed peripheral blood mononuclear cells were lentivirally transduced with the CD123CAR vector. Prior to T cell infusion, all patients undergo a lymphodepleting regimen including fludarabine 25-30 mg/m2 daily for 3 days and cyclophosphamide 300 mg/m2 daily for 3 days. Pts receive a single dose of CD123CAR T cells with an option for a second infusion if they continue to meet safety and eligibility criteria and still have CD123+ disease. To date, 14 patients have been enrolled and 7 treated (6 AML, 1 BPDCN). All 6 patients in the AML cohort had refractory AML following alloHSCT, and a median of 4 (range: 4-7) prior lines of therapy. Of the 2 pts treated at dose level (DL) 1 (50M CAR+ T), 1 achieved a morphologic leukemic-free state, which lasted 2 months. She received a second infusion 3 months later with subsequent blast reduction from 77.9% to 0.9% (flow cytometry) after 35 days. Of the 4 pts on DL 2 (200M CAR+ T), 1 achieved CR and became transfusion independent. She proceeded to a second alloHSCT on day 70. Restaging on day +161 post-transplant showed she has remained in MRD-negative CR with good engraftment and 100% donor chimerism. Another pt with CR prior to treatment remained in CR at day 28 and has proceeded to a second alloHSCT. The remaining 2 patients had reductions in blast counts, but did not achieve remission. All toxicities were reversible and manageable: cytokine release syndrome (CRS; 4 grade 1, 1 grade 2); 1 adenoviral pneumonia requiring intubation; and 1 grade 3 rash due to drug hypersensitivity. There were no dose limiting toxicities and no treatment-related cytopenias. One pt with prior alloHSCT had mild recurrent cutaneous GVHD, which occurred after the completion of CAR T treatment. Correlative studies including T cell persistence and CD123 expression are underway and will be reported. In the BPDCN cohort, 1 pt has been treated so far, a 74-year-old man with a persistent bulky subcutaneous mass after clinical trial treatment with a CD123 antibody-drug conjugate. Following lymphodepletion, he received a single dose of 100M autologous CD123CAR T cells and continues to be in CR at 60 days post-infusion. Skin biopsies at the tumor site on days 14 and 28 showed no evidence of disease. Restaging work-up at day 28 revealed disease-free bone marrow, no new masses by CT scan, normalized blood counts, and complete resolution of clinical symptoms. The pt tolerated the treatment well with no CRS or neurologic toxicity. In this first-in-human clinical trial of CD123CAR T cell therapy, we have demonstrated the feasibility and safety of targeting CD123. We have also observed promising anti-leukemic activity in both AML and BPDCN. Importantly, no myeloablative effects have been observed, supporting further testing of this immunotherapeutic strategy in both transplant eligible and ineligible patients. Disclosures Stein: Stemline: Consultancy; Amgen: Consultancy, Speakers Bureau.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 13-13 ◽  
Author(s):  
Chaoke Bu ◽  
Zhiyong Peng ◽  
Min Luo ◽  
Guangchao Li ◽  
Chunfu Li

Background Chimeric antigen receptor (CAR)-T cell therapy has demonstrated remarkable success in treating a variety of blood cancers, such as CD19 CAR-T for B-cell malignancies and BCMA CAR-T for myeloid myeloma (MM). However, similar progress has yet to be achieved with relapsed and refractory acute myeloid leukemia (R/R AML), primarily due to the heterogeneous nature of AML, making it difficult to find an ideal CAR-T target. Previous efforts have targeted single CD33, CD123, LeY, NKG2D, or CD70 receptors, but the overall response rate is much lower compared to CD19 CAR-T. Improved clinical outcome was recently reported with a dual-receptor CAR-T (CD33 and CLL1), reaching ~80% CR for R/R AML patients. However, CD33 is also expressed in normal hematopoietic stem cells (HSC) and patients need allogeneic hematopoietic stem cell transplantation (HSCT) following CAR-T therapy. Notably, AML mainly affects the elder population, and patients more than 65 years old are usually not suitable for HSCT. To address these challenges, we aim to find an effective target for AML without the need for the HSCT. In our study, CLL1 is chosen as a promising target as it is not expressed on normal HSCs, but highly expressed on AML blasts cells and leukemia stem cells (LSCs), which is a small population that plays an important role in disease progression and relapse. Here we report the results from a Phase I clinical trial to evaluate the toxicity and efficacy of the CLL1 CAR-T in the treatment of pediatric R/R AML. Methods We have generated a 2nd generation of CLL1 CAR-T, the extracellular scFv was derived from a murine CLL1 monoclonal antibody, which was generated by hybridoma technology. Autologous CAR-T cells were manufactured in a cGMP facility. Between Oct 2019 and Mar 2020, 3 pediatric R/R AML patients were infused. CAR-T cells were given by a dose at 0.2-1x106/kg with a single dose. Results Of the 3 patients infused, cytokine release syndrome (CRS) occurred in 3 patients (2 grade Ⅰ, 1grade II), no neurotoxicity occurred. All patients suffered pancytopenia, granulocytopenia and monocytopenia. All the adverse effects were resolved after treatment. Patient 1 is a refractory AML patient with more than 95% of AML blasts being CLL1 positive. The infused CAR-T cells showed typical clonal expansion peaking at Day 8. The patient reached CR/MRD- when evaluated at Day 21 post infusion. The patient received HSCT at Day 35, and remained CR/MRD- till now (Jul 2020, 8 months post CAR-T infusion). Patient 2 is a relapsed patient after HSCT and showed severe bone marrow necrosis (BMN). Therefore, we only generated a dose 0.35-1x106/kg CAR-T cells. The patient reached CR/MRD- when evaluated at Day 14 post infusion. The patient went through a second HSCT at Day 38. However, the patient passed away due to GVHD two months after the second HSCT. Patient 3 is a refractory AML patient with about 85% of AML blasts being CLL1 positive. The patient reached CR/MRD+ (1%) when evaluated at Day 14 post infusion. However, AML blast increased to 11% when evaluated at Day 30, a majority of them were CLL1 negative. The patient received Azacitidine treatment at Day 30-34, and AML blast decreased to 2.8% at Day 45. Notably this patient was refractory to Azacitidine treatment previously, suggesting that CLL1 CAR-T could eradicate LSCs, making the left AML tumor cells sensitive to chemotherapy. This patient received HSCT at Day 75, and remained CR/MRD- till now (Jul 2020, 7 months post CAR-T infusion). Conclusion Our study suggested that CLL1 CAR-T is a therapy with high efficacy and manageable toxicity in R/R AML patients. All patients in this study could reach CR within one month and only experienced 1-2 grade CRS. For patients with CLL1 negative AML blast, the CLL1- cells may take over after CLL1 CAR-T therapy. Combination with chemotherapy like Azacitidine may help patients reach complete response. Figure Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Author(s):  
Martin Wermke ◽  
Sabrina Kraus ◽  
Armin Ehninger ◽  
Ralf C. Bargou ◽  
Maria-Elisabeth Goebeler ◽  
...  

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4825-4825
Author(s):  
Moazzam Shahzad ◽  
Sibgha Gull Chaudhary ◽  
Ali Hussain ◽  
Fatima Ali ◽  
Ayesha Khalid ◽  
...  

Abstract Background: Acute myeloid leukemia (AML) is a clonal hematologic malignancy that generally affects older adults. Despite achieving complete remission (CR) in over two-third of patients with initial induction therapy and subsequent allogeneic hematopoietic stem cell transplantation in intermediate-high risk patients, more than half of AML patients experience disease relapse. The prognosis of patients with relapsed/refractory AML (RR-AML) is often poor and treatment modalities are limited. Chimeric antigen receptor T cell (CAR-T) therapy has shown promising results in lymphoid malignancies and myeloma, and these are now being explored for the management of RR-AML. In this systematic review and meta-analysis, we aimed to investigate the outcomes of CAR-T therapy in RR-AML patients. Methods: Following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines, a comprehensive literature search was conducted on three databases (PubMed, Cochrane Register of Controlled Trials, and Clinical trials.gov) using MeSH terms and keywords for "Leukemia, Myeloid, Acute" AND "Receptors, Chimeric Antigen" OR "adoptive immunotherapy" from the date of inception to April 2021. A total of 673 articles were screened and original studies reporting patients with RR-AML having CAR-T therapy as the only intervention were included while reviews, duplicate, and non-relevant articles were excluded. A total of 10 studies (8 clinical trials and 2 case reports) were included. The data for following outcomes were extracted: complete response (CR), partial response (PR), overall response rate (ORR), overall survival (OS), progression-free survival (PFS), stable disease (SD), progressive disease (PD), cytokine release syndrome (CRS) and neurotoxicity (NT). Quality evaluation was done using the NIH quality assessment tool. Inter-study variance was calculated using the Der Simonian-Laird Estimator. Proportions along with 95% confidence Interval (CI) were extracted to compute pooled analysis using the 'meta' package by Schwarzer et al. in the R programming language (version 4.16-2). Results: We identified 39 patients in 10 studies who received CAR-T therapy for RR-AML. Median age of patients was 35 (7.3-80) years and 59% (n=23) were male. The median follow-up time was 5 (0.7-23) months. (Table 1) Four patients had history of allogeneic hematopoietic stem cell transplant (HCT) prior to CAR-T therapy while subsequent HCT was performed in 5 patients. The pooled analysis showed a CR and ORR of 38.5% (95% CI 0.03-0.81, I 2 =66%, n=29) and 56% (CI 0.18-0.91, I 2=58%, n=29), respectively. Median duration of response was 5.5 (1-23) months. OS was reported from 1.9 months to 23 months. The pooled incidence of CRS and NT were 42.7% (95%CI 0.06-0.87, I 2=66%, n=28) and 1.3% (95% CI 0.00-0.16, I 2= 0%, n=21) respectively. Graft-versus-host disease (GVHD) was reported in 2 patients who had prior and subsequent HCT after CAR-T therapy; first patient developed grade IV GVHD in the setting of salvage therapy with donor lymphocyte infusions for relapsed disease 6 months' post CAR-T and 4 months post second allo-HCT while second patient received CAR-T as part of conditioning therapy and developed grade IV GVHD on day 32. Conclusion: CAR-T therapy has shown favorable results comparable to current salvage therapies for relapsed or refractory AML with an acceptable toxicity profile. However, there are several challenges including the heterogeneous biology of AML, lack of a targetable antigen expression on malignant cells, and immune escape and exhaustion. Future prospective studies with improved CAR-T constructs will hopefully improve the outcomes in this therapeutically challenging patient population. Figure 1 Figure 1. Disclosures Lin: AbbVie, Aptevo Therapeutics, Astellas Pharma, Bio-Path Holdings, Celgene, Celyad, Genentech-Roche, Gilead Sciences, Incyte, Jazz Pharmaceuticals, Novartis, Ono Pharmaceutical, Pfizer, Prescient Therapeutics, Seattle Genetics, Tolero, Trovagene: Research Funding. Abhyankar: Incyte/Therakos: Consultancy, Research Funding, Speakers Bureau. McGuirk: EcoR1 Capital: Consultancy; Juno Therapeutics: Consultancy, Honoraria, Research Funding; Pluristem Therapeutics: Research Funding; Bellicum Pharmaceuticals: Research Funding; Gamida Cell: Research Funding; Novartis: Research Funding; Astelllas Pharma: Research Funding; Kite/ Gilead: Consultancy, Honoraria, Other: travel accommodations, expense, Kite a Gilead company, Research Funding, Speakers Bureau; Fresenius Biotech: Research Funding; Novartis: Research Funding; Magenta Therapeutics: Consultancy, Honoraria, Research Funding; Allovir: Consultancy, Honoraria, Research Funding.


Sign in / Sign up

Export Citation Format

Share Document