scholarly journals A Novel Chimeric Antigen Receptor T Cells Therapy Strategy That Dual Targeting CD19 and CD123 to Treat Relapsed Acute Lymphoblastic Leukemia after Allogeneic Hematopoietic Stem Cell Transplantation

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4015-4015 ◽  
Author(s):  
Sanfang Tu ◽  
Lan Deng ◽  
Rui Huang ◽  
Xuan Zhou ◽  
Jilong Yang ◽  
...  

Abstract BACKGROUND: The prognosis of patients with relapsed B-lineage acute lymphocyte leukemia (B-ALL) after allogeneic hematopoietic stem cells transplantation(HSCT) is poor. It is difficult to obtain disease-free remission and long-term survival in these patients based on chemotherapy, donor leukocyte infusion (DLI) or molecular targeted therapy. Donor-derived anti-CD19 CAR-T(CAR 19) cells may obtain remission in these patients, but many still encounter relapse, especially CD19-negative relapse. CD123 is a surface marker not only associated with the progenitors of acute myeloid leukemia, but also found in progenitors of acute lymphoblastic leukemia. Recent studies have shown that CD123 is highly expressed in the patients with CD19-negative relapsed after CAR19 cells therapy. we developed a novel therapeutic strategy for to administer pooled donor-derived fourth generation CART cells targeting CD19 and CD123 respectively, to patients who relapsed following allogeneic HSCT to avoid CD19-negative relapse. In this study, we report three patients who received combination CARTs therapy achieved disease-free survival that at least more than 6 months (www.clinicaltrials.gov; #NCT03125577). PATIENTS AND METHODS: Three patients with relapsed B-ALL after HLA-matched sibling HSCT have been enrolled in the study to date, and all of their leukemia cells highly expressed CD19 and CD123 antigens. The first and second patients experienced relapse 6 months and one year after all-HSCT respectively; their B-ALLs were p190-positive and carried T315I mutation, and resisted to bonatinib, chemotherapy and DLI. The third patient was also refractory to chemotherapy and DLI. All patients received fludarabine (FLU) and cyclophosphamide (CTX) conditioning chemotherapy (FLU 30mg/m2, d1-3; CTX 300mg/m2, d1-3) before CART infusions. Donor T cells were apheresis collected and transduced with an apoptosis-inducible, safety-engineered lentiviral CD19 or CD123 scFv CAR fused with intracellular signaling domains: CD28/CD27/CD3ζ-iCasp9 (4SCAR19 and 4SCAR123). CAR-T cells were infused at dose range of 0.26-1.38x106 cells/kg. The quality of apheresis cells, gene transfer and T cell proliferation efficiencies, and effective CAR T infusion dose were quantitatively scored and documented. RESULTS: All three patients achieved minimal residual disease (MRD) negative remission within 1 month after CAR-T infusions. Monthly follow-ups of the first and second patients indicated that they achieved stable MRD-negative and p190-negative remission, and remained disease-free for 7 months and 11 months, respectively. The third patient was MRD-positive but achieved bone marrow morphological remission at 7 month follow-up time. Flow cytometry analysis of the MRD cells detected CD19 positive and CD123 partial positive ALL clones, and note that the third patient received the lowest dose of CART infusion, 0.26x10e6/kg. The first patient developed grade 1 cytokine release syndrome (CRS) after CAR-T cell infusions. The second patient developed grade 1 oral acute graft-versus-host disease (aGVHD) and pulmonary infection. The third patient developed grade 2 CRS, with hypoxemia and unilateral massive pleural effusion. We detected high IL-6 in his pleural fluid >5000 IU/L, but serum level IL-6 is normal. There was no significant absorption of pleural effusion after treatment with anti-interleukin 6 receptor monoclonal antibody. The patient improved after 5 days of treatment with chest drainage and dexamethasone 10mg/qd. None of the three patients developed central nervous system toxicity, and there was no greater than grade 2 CRS and severe myelosuppression, consistent with the safety profile of the 4SCAR design. CONCLUSIONS: We have successfully treated three relapsed allo-HSCT B-ALL patients using donor-derived 4SCAR19 and 4SCAR123 T cells. All three patients achieved long-term disease-free survival without severe CRS and GVHD. Thus, the administration of double 4SCAR19/4SCAR123 T cells may overcome CD19 escape and prolong disease-free survival. Disclosures No relevant conflicts of interest to declare.

1989 ◽  
Vol 7 (6) ◽  
pp. 747-753 ◽  
Author(s):  
P Bordigoni ◽  
J P Vernant ◽  
G Souillet ◽  
E Gluckman ◽  
D Marininchi ◽  
...  

Thirty-two children ranging in age from 1.5 to 16 years with poor-prognosis acute lymphoblastic leukemia (ALL) were treated with myeloablative immunosuppressive therapy consisting of cyclophosphamide (CPM) and total body irradiation (TBI) followed by allogeneic bone marrow transplantation (BMT) while in first complete remission (CR). The main reasons for assignment to BMT were WBC count greater than 100,000/microL, structural chromosomal abnormalities, and resistance to initial induction therapy. All children were transplanted with marrow from histocompatible siblings. Twenty-seven patients are alive in first CR for 7 to 82 months post-transplantation (median, 30 months). The actuarial disease-free survival rate is 84.4% (confidence interval, 7.2% to 29%) and the actuarial relapse rate is 3.5% (confidence interval, 0.9% to 13%). Four patients died of transplant-related complications, 16 developed low-grade acute graft-v-host disease (GVHD), and six developed chronic GVHD. The very low incidence of relapse (one of 28 long-term survivors) precluded the determination of the prognostic significance of the different poor-outcome features. Moreover, two infants treated with busulfan, CPM, and cytarabine (Ara-C) relapsed promptly in the marrow. In summary, as a means of providing long-term disease-free survival and possible cure, BMT should be considered for children with ALL presenting poor-prognostic features, particularly certain chromosomal translocations [t(4;11), t(9;22)], very high WBC counts, notably if associated with a non-T immunophenotype, and, perhaps, a poor response to initial therapy with corticosteroids (CS), or infants less than 6 months of age.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2696-2696 ◽  
Author(s):  
Sanfang Tu ◽  
Rui Huang ◽  
Lan Deng ◽  
Xuan Zhou ◽  
Yanjie He ◽  
...  

Abstract Background: CD19 targeting chimeric antigen receptor T cells(CART19) therapy have shown great therapeutic potential in relapsed and refractory B cell acute lymphoblastic leukemia (ALL), but associated with risk of life-threatening adverse effects as severe cytokine release syndrome (sCRS) and CAR-T-cell-related encephalopathy syndrome (CRES).It's been reported that high leukemia burden before CART therapy, and high does infused CART cells are associated with severity of CRS and CRES. To decrease the risk of severe adverse effect, we applied integrated therapeutic strategy of using fourth generation CART cells , reducing leukemia blasts burden in bone marrow, and decreasing the dose of infused CART cells (www.clinicaltrials.gov; #NCT03407859 and #NCT03125577 ). Methods: Between May 23, 2016 and July 2, 2018,the trial enrolled 20 patients (pts) who were exhausted with all available treatment options, life expectancy >2 months,CD19-positive and diagnosis of B lineage ALL.T cells were apheresis collected and transduced with an apoptosis-inducible, safety-engineered lentivector CAR with the following intracellular signaling domains: CD28/CD27/CD3ζ-iCasp9 (4SCAR). In vitro amplication of CART was not performed. In pts with bone marrow blasts exceeding 50.0%, VDCP or similar chemotherapy was given to reduce the tumor burden, and then received FC conditioning regimen (FLU 30mg/m2, d1-3; CTX 300mg/m2, d1-3), while FC regimen was directly carried out in pts with bone marrow blasts less than 50.0%.In this trial, Pts received single CD19-targeted CARTs or multi-CARTs targeting CD19 and an additional antigen of CD22 or CD123.The level of CAR-T cells and cytokines in peripheral blood, as well as tumor burden was measured after infusion. Results: 20 pts were enrolled and infused with CAR-T cells. The median age is 37.5 (16-67) years old. Of these pts, 5 had prior HSCT and 14 had adverse genetic abnormalities, including 4 pts (20%) who were Philadelphia chromosome-positive(Ph+). All pts were previously treated with 2-22 courses of cytotoxic chemotherapy regimens. The overall objective response rate was 85%(17/20), and the complete response(CR) rate was 80%(16/20). The complete remission rate of 12 pts receiving single CD19-targeted CART therapy was 83 %(10/12), while 33%(4/12) of them had disease relapse at 6 months after infusion. Of the 7 people who received multi-CARTs infusion, 71%(5/7) achieved complete remission, with relapse rate of 29%(2/7) at 6 months after infusion. 3 pts who relapsed post transplantation received combination therapy of anti-CD19 CART and anti-CD123 CART, and all achieved minimal residual disease-negative CR within 1 month after CART infusion, 2 of whom maintained disease-free survival for 7 months and 11 months to date, respectively. Among the 7 people who underwent HSCT after achieving CR, 6 of them maintained disease-free survival for 3 months to 9 months. At a median follow-up of 115.5 days (ranging from14 to 384), the median overall survival was 269 days and the median event-free survival was 232 days. During treatment and follow-up, the most common adverse events were grade 1-2 cytokine release syndrome (CRS), with an incidence of 55%.No grade 3 or higher CRS was observed.12 pts were infused with a dose equal to or exceeding 5*10^5/kg, 10 of whom had CRS response. While only 2 of the 8 pts who received the infusion dose of less than 5*10^5/kg had CRS reaction and both of them were grade 1 CRS, suggesting low CAR-T cell doses decrease the risk of CRS (P=0.009). Interestingly, the objective response rate did not differ significantly between the low dose and high dose group. Conclusions: Based on fourth generation CART system, a therapeutic strategy of low tumor burden and low CART infusion dose shows a safer profiling while remaining potent efficacy against leukemia. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2126-2126 ◽  
Author(s):  
Shuangyou Liu ◽  
Biping Deng ◽  
Yuehui Lin ◽  
Zhichao Yin ◽  
Jing Pan ◽  
...  

Abstract With traditional therapies, the prognosis of relapsed acute lymphoblastic leukemia (ALL) after allogeneic hematopoietic stem cell transplantation (allo-HSCT) is extremely poor. Chimeric antigen receptor (CAR) T cell therapy targeting at CD19 has demonstrated a significant efficacy on refractory/relapsed (r/r) B-ALL, but single-target CART could not maintain a long-term remission. Recently, CD22-CART has also shown an exciting result in r/r B-ALL. Here we sequentially applied CD19- and CD22-specific CART cells to treat relapsed B-ALL post-HSCT and observed the therapeutic effect. From June 30,2017 through May 31,2018, twenty-four B-ALL patients (pts) relapsing after allo-HSCT with both antigens CD19 and CD22 expression on blasts were enrolled, the median age was 24 (2.3-55) years. Seventeen pts had hematologic relapse, 6 with both bone marrow and extramedullary (EM) involvements and 1 with EM disease (EMD) only. Fourteen pts had failed to previous therapies including chemotherapy, donor lymphocyte infusion, interferon and even murinized CD19-CART in other hospitals. Recipient-derived donor T cells were collected for producing CAR-T cells, which were transfected by a lentiviral vector encoding the CAR composed of CD3ζ and 4-1BB. Eighteen pts were initially infused with murinized CD19-CART, then humanized CD22-CART; while 6 pts (5 failed to prior murinized CD19-CART and 1 had bright CD22-expression) were initially infused with humanized CD22-CART, then humanized CD19-CART. The time interval between two infusions was 1.5-6 months based on patients' clinical conditions. The average dose of infused CAR T cells was 1.4×105/kg (0.4-9.2×105/kg) for CD19 and 1.9×105/kg (0.55-6.6×105/kg) for CD22. All patients received fludarabine with or without cyclophosphamide prior to each infusion, some pts accepted additional chemo drugs to reduce the disease burden. Treatment effects were evaluated on day 30 and then monthly after each CART, minimal residual disease (MRD) was detected by flow cytometry (FCM) and quantitative PCR for fusion genes, EMD was examined by PET-CT, CT or MRI. Sixteen patients finished sequential CD19- and CD22-CART therapies. Three cases could not undergo the second round of CART infusion (1 died, 1 gave up and 1 developed extensive chronic graft-versus-host disease (GVHD)). The rest of 5 pts are waiting for the second CART. After first T-cell infusion, 20/24 (83.3%) pts achieved complete remission (CR) or CR with incomplete count recovery (CRi), MRD-negative was 100% in CR or CRi pts, 3 (12.5%) cases with multiple EMD obtained partial remission (PR), and 1 (4.2%) died of severe cytokine release syndrome (CRS) and severe acute hepatic GVHD. Sixteen patients (15 CR and 1 PR) underwent the second CART therapy. Before second infusion, 3/15 pts in CR became MRD+ and others remained MRD-. On day 30 post-infusion, 1 of 3 MRD+ pts turned to MRD-, 1 maintained MRD+ ( BCR/ABL+) and 1 had no response then hematologic relapse later. The PR patient still had not obtained CR and then disease progressed. As of 31 May 2018, at a median follow-up of 6.5 (4-10) months, among 16 pts who received sequential CD-19 and CD-22 CART therapies, 1 had disease progression, 2 presented with hematological relapse and 2 with BCR/ABL+ only, the overall survival (OS) rate was 100% (16/16), disease-free survival (DFS) was 81.3% (13/16) and MRD-free survival was 68.8% (11/16). CRS occurred in 91.7% (22/24) pts in the first round of T-cell infusion, most of them were mild-moderate (grade I-II), merely 2 pts experienced severe CRS (grade III-IV). The second CART only caused grade I or no CRS since the leukemia burden was very low. GVHD induced by CART therapy was a major adverse event in these post-HSCT patients. After the first CART, 7/24 (29.2%) pts experienced GVHD, of them, 4 presented with mild skin GVHD, 2 with severe hepatic GVHD (1 recovered and 1 died), and 1 developed extensive chronic GVHD. No severe GVHD occurred in the second infusion. Our preliminary clinical study showed that for B-ALL patients who relapsed after allo-HSCT, single CD19- or CD22- CART infusion resulted in a high CR rate of 83.3%, sequentially combined CD19- and CD22-CART therapies significantly improved treatment outcome with the rate of OS, DFS and MRD-free survival being 100%, 81.3% and 68.8%, respectively, at a median follow-up of 6.5 months. The effect of CART on multiple EMD was not good and CART induced GVHD needs to be cautious. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1753-1753
Author(s):  
Wei Cui ◽  
Xinyue Zhang ◽  
Haiping Dai ◽  
Qingya Cui ◽  
Jia Yin ◽  
...  

Abstract Background: CD19 chimeric antigen receptor T (CAR-T) cells have demonstrated impressive early response rates in relapse and refractory B acute lymphoblastic leukemia (r/r B-ALL). However, a high rate of patients suffered a relapse, which occurred in a large subset of other trials and confers dismal outcomes. Dual targets approaches are proved to optimize the response rate and prevent CD19 - relapse. Alternatively, limited patients accepted the tandem CD19/CD22 CAR-T therapy, but the clinical trials in a large scale to investigate the tandem CAR-T were rare. Methods: We conducted an open label, single center clinical trial at the First Affiliated Hospital of Soochow University to investigate the efficacy and safety of tandem CD19/CD22 dual targets CAR T-cells for r/r B-ALL. All patients received FC (fludarabine, 30 mg/m2, days 1-3 and cyclophosphamide, 300 mg/m2, days 1-3) based chemotherapy as the lymphodepleting regimen. Median infusion dose of CAR-T cells was 1(0.5-2.5) *10 7 cells/kg. Results: From October 2017 to June 2021, a total of 47 patients were treated with CD19/CD22 CAR T-cells and included in our analysis. Among the 47 patients, primary refractory B-ALL patients account for 44.68%. 27 patients (57.4%) had a high disease burden, with 20% or more blasts in BM. Consequently, at day 28 assessment, 47 patients (100%) got hematological CR and the 40 out of 47 patients (85.1%) achieved minimal residual disease complete remission (MRD -CR). The toxicities of CD19/CD22 CAR T-cells therapy were reversible and clinically manageable. Cytokine reverse syndrome of any grades occurred in 41 of 47 patients (87.23%) and was severe (grade>2) in 8 (17.02%). Immune effector cell-associated neurotoxicity happened in one patient. The most common severe hematological abnormalities were grade 3/4 leukopenia (74.47%). Serious thrombocytopenia and anemia occurred in 48.93% and 57.44% patients. The non-hematological toxicity were reversible with tight monitoring and support care. Within a median follow-up of 21.83 months (range, 2.57 to 42.53), the median overall survival (OS) and leukemia free survival (LFS) for the entire cohort have not reached. The OS rate of all the patients was 93.569% (95%CI, 80.97% to 97.832%) at 6 months, 78.721% (95%CI, 60.719% to 87.625%) at 1 year and 74.578% (95%CI, 55.263% to 84.969%) at 2 years in all patients. The LFS rate was 87.031% (95%CI, 73.375% to 93.958%) and 68.297% (95%CI, 51.419% to 80.365%) at 6 month and 1 year, respectively. The 6-months cumulative incidence of relapse (CIR) was 8.96%, while 1-year CIR was 23.254%. Thirty-four of 47 patients (72.34%) proceeded to a bridging allogeneic hematopoietic stem cell transplantation (allo-HSCT). The OS of HSCT group was 94.118% at 6 months and 80.420% at 1 year. The 6-months OS of no-HSCT group was 83.916%, while 1-year OS was 74.592%. The HSCT group had significantly better LFS and lower CIR than the no-HSCT group (LFS, p=0.0459; CIR, p=0.0267). We initially performed multivariable Cox regression analyses, which shows that better long-term survival in patients with MRD -CR status, as well as bridging allo-HSCT. Conclusions: Tandem CD19/CD22 CAR-T cells are safety and highly effective in inducing CR for r/r B-ALL patients. The consolidative allo-HSCT can provide long-term durable disease control in these patients. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5389-5389 ◽  
Author(s):  
Sanfang Tu ◽  
Rui Huang ◽  
Xuan Zhou ◽  
Yanjie He ◽  
Zhao Liang ◽  
...  

Abstract Background: Chimeric antigen receptor-modified T cells (CARTs) targeting CD19 has illustrated therapeutic efficacy in diffuse large B cell lymphoma (DLBCL). However, severe cytokine release syndrome (sCRS) reaction and CAR-T-cell-related encephalopathy syndrome (CRES) are the main adverse reactions of CART therapy. Because of the concern of severe CRES which is associated with potential mortality, patients with primary central nervous system Lymphoma(PCNSL) are excluded in most of CART clinical trials. CD70 is a promising therapeutic target due to the restricted expression pattern in normal tissues and overexpression in some lymphoma tissues, so CD70 specific CARTs maybe avoid CD19-negative relapse .Here we report a case with refractory and relapsed PCNSL who achieved long-term disease-free survival by combination therapy of CD19 and CD70 specific CARTs without inducing CRS or CRES. Methods: CD3+ cells were selected from the apheresis PBMC and activated before lentiviral 4SCAR infection. The cells were transduced with a caspase 9-inducible, safety-engineered lentivector CAR containing anti-CD19 or -CD70 scFv fused with multiple intracellular signaling domains: CD28/CD27/CD3z-iCasp9 (4SCAR19 or 4SCAR70). The quality of apheresis cells, gene transfer and T cell proliferation efficiencies, and effective CAR T infusion dose were quantitatively scored and documented. Tumor sections were immunohistochemically stained with different antibodies including anti-CD19 and anti-CD70 antibodies to confirm antigen expression. The patient received cyclophosphamide and fludarabine conditioning regimen before the dual CARTs cell infusions. Case presentation: A 67-year-old male who was diagnosed as PCNSL of DLBCL in 2011.The patient received 6 cycles of temozolomide and high does methotrexate, and achieved CR. In December 2016 he relapsed and was treated with right frontal lobe space-occupying resection and multi-course chemotherapy including 2 course of rituximab, high dose methotrexate and temozolomide ,and 6 courses of rituximab, high dose methotrexate and ibrutinib,and he achieved remission again.However, He relapsed again in August 2017, with the clinical symptoms of dizziness , fatigue, left corners of the mouth askew and occlusal difficulties.MRI presented a residual mass on the right side of the postoperative cavity of 26mm*35mm*30mm and stale hemorrhage in left basal ganglia. After confirmation for CD19 and CD70 expression in his tumor specimens, the pt enrolled in the clinicaltrail :"Combination CART cell therapy targeting hematological malignancies"(clinicaltrail.gov registry NCT03125577).He received 4SCAR19 and 4SCAR70 T cells targeting CD19 and CD70 following lymphodepletion chemotherapy with FC regimen conditioning (FLU 30mg/m2, d1-3; CTX 300mg/m2, d1-3) in October 2017. Brain MRI 1 month later showed complete remission and the pt had symptomatic improvement. To date, after more than 8 months of follow-up, the pt remains in CR (figure1) and CART cells still can be detected . There was no CRS or CRES during the treatment and in follow-up period. Conclusions: The study results demonstrate that CARTs are able to pass through the blood-brain barrier without inducing CRS or CRES, suggesting that CNS tumor is not an absolute contraindication to 4sCART therapy. In addition, the combination of CD19 and CD70 specific 4sCARTs could effectively target PCNSL and achieves long-term disease-free survival without sCRS or CRES. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1991 ◽  
Vol 78 (11) ◽  
pp. 2814-2822 ◽  
Author(s):  
CA Linker ◽  
LJ Levitt ◽  
M O'Donnell ◽  
SJ Forman ◽  
CA Ries

Abstract We treated 109 patients with adult acute lymphoblastic leukemia (ALL) diagnosed by histochemical and immunologic techniques. Patients were excluded only for age greater than 50 years and Burkitt's leukemia. Treatment included a four-drug remission induction phase followed by alternating cycles of noncrossresistant chemotherapy and prolonged oral maintenance therapy. Eighty-eight percent of patients entered complete remission. With a median follow-up of 77 months (range, 48 to 111 months), 42% +/- 6% (SEM) of patients achieving remission are projected to remain disease-free at 5 years, and disease-free survival for all patients entered on study is 35% +/- 5%. Failure to achieve remission within the first 4 weeks of therapy and the presence of the Philadelphia chromosome are associated with a 100% risk of relapse. Remission patients with neither of these adverse features have a 48% +/- 6% probability of remaining in continuous remission for 5 years. Patients with T-cell phenotype have a favorable prognosis with 59% +/- 13% of patients achieving remission remaining disease-free compared with 31% +/- 7% of CALLA-positive patients. Intensive chemotherapy may produce prolonged disease-free survival in a sizable fraction of adults with ALL. Improved therapy is needed, especially for patients with adverse prognostic features.


Sign in / Sign up

Export Citation Format

Share Document