Targeted Therapy With the T-Cell–Engaging Antibody Blinatumomab of Chemotherapy-Refractory Minimal Residual Disease in B-Lineage Acute Lymphoblastic Leukemia Patients Results in High Response Rate and Prolonged Leukemia-Free Survival

2011 ◽  
Vol 29 (18) ◽  
pp. 2493-2498 ◽  
Author(s):  
Max S. Topp ◽  
Peter Kufer ◽  
Nicola Gökbuget ◽  
Mariele Goebeler ◽  
Matthias Klinger ◽  
...  

Purpose Blinatumomab, a bispecific single-chain antibody targeting the CD19 antigen, is a member of a novel class of antibodies that redirect T cells for selective lysis of tumor cells. In acute lymphoblastic leukemia (ALL), persistence or relapse of minimal residual disease (MRD) after chemotherapy indicates resistance to chemotherapy and results in hematologic relapse. A phase II clinical study was conducted to determine the efficacy of blinatumomab in MRD-positive B-lineage ALL. Patients and Methods Patients with MRD persistence or relapse after induction and consolidation therapy were included. MRD was assessed by quantitative reverse transcriptase polymerase chain reaction for either rearrangements of immunoglobulin or T-cell receptor genes, or specific genetic aberrations. Blinatumomab was administered as a 4-week continuous intravenous infusion at a dose of 15 μg/m2/24 hours. Results Twenty-one patients were treated, of whom 16 patients became MRD negative. One patient was not evaluable due to a grade 3 adverse event leading to treatment discontinuation. Among the 16 responders, 12 patients had been molecularly refractory to previous chemotherapy. Probability for relapse-free survival is 78% at a median follow-up of 405 days. The most frequent grade 3 and 4 adverse event was lymphopenia, which was completely reversible like most other adverse events. Conclusion Blinatumomab is an efficacious and well-tolerated treatment in patients with MRD-positive B-lineage ALL after intensive chemotherapy. T cells engaged by blinatumomab seem capable of eradicating chemotherapy-resistant tumor cells that otherwise cause clinical relapse.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 174-174 ◽  
Author(s):  
Max S Topp ◽  
Gerhard Zugmaier ◽  
Nicola Goekbuget ◽  
Svenja Neumann ◽  
Heinz-August Horst ◽  
...  

Abstract Abstract 174 Blinatumomab, a bispecific, T cell-engaging (BiTE®) antibody, can effectively redirect T cells for highly selective lysis of CD19+ target cells. The B-cell differentiation antigen CD19 is a marker for B-ALL cells. In B-lineage acute lymphoblastic leukemia (ALL), persistence or relapse of minimal residual disease (MRD) is an independent poor prognostic factor, and new treatments are urgently needed. MRD relapse during or after maintenance treatment in adult standard risk patients generally heralds a hematological relapse in 90% of patients. A phase 2 study was conducted to determine the efficacy of blinatumomab in ALL patients with MRD persistence or relapse (MRD level <310-4) after induction and consolidation therapy. MRD was assessed by qRT-PCR for rearrangements of immunoglobulin (Ig) or T-cell receptor (TCR) genes, or for specific fusion genes. Blinatumomab was administered as a 4-week continuous i.v. infusion at a dose of 15 μ g/m2/d followed by a 2-week treatment free period (1 cycle). Primary endpoint was the proportion of patients with MRD response defined by individual rearrarrangement of Ig or TCR below 10-4. Patients who showed neither MRD progression nor response were permitted to receive up to 7 cycles of treatment. Patients who had achieved MRD response received 3 additional consolidation cycles. Between May 2008 and November 2009, 21 patients (16 Ph-negative; 2 patients with MLL-AF4; 5 patients with Ph+ ALL) were enrolled. The cut-off date for data analysis was May 15, 2010. Patients received between 1 and 7 cycles of blinatumomab (total of 66 cycles). Transient pyrexia (100%) and chills (43%) were the most common clinical AEs. There were no blinatumomab related deaths. Sixteen patients became MRD-negative. One patient was not evaluable due to a grade 3 adverse event (AE) leading to treatment discontinuation. Of the responding patients, 13 had never before achieved a negative MRD status on chemotherapy. Regardless of their MRD level prior to study treatment, all 16 (13/15 patients with Ph− and 3/5 patients with Ph+ ALL) became MRD-negative after the first cycle of blinatumomab. Nine patients were enrolled with a MRD load >10-2 prior to study treatment and all reached complete MRD response. Thirteen out of 16 patients with persisting MRD prior to study treatment and 3 out of 4 patients with MRD relapse showed complete MRD response. Overall relapse-free survival (RFS) currently is 78% at a median follow up of 405 days. RFS is 100% for the 8 patients who received subsequent allogeneic stem cell transplantation (median follow up 434 days). Blinatumomab is a highly active treatment for patients with MRD-positive B-lineage ALL after intensive chemotherapy and has an acceptable safety profile. T cells engaged by blinatumomab seem capable of eradicating chemotherapy-resistant tumor cells in bone marrow that otherwise might cause clinical relapse. A long RFS suggests that blinatumomab may improve outcome in patients with B-precursor ALL. A multicenter international study of blinatumomab in patients with MRD-positive B-lineage ALL has been initiated. Disclosures: Zugmaier: Micromet Inc.: Employment. Degenhard:Micromet Inc.: Employment. Schmidt:Micromet Inc.: Employment. Scheele:Micromet Inc.: Employment. Kufer:Micromet Inc.: Employment. Klinger:Micromet Inc.: Employment. Nagorsen:Micromet Inc.: Employment. Bargou:Micromet Inc.: Consultancy.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Stephanie L. Rellick ◽  
Gangqing Hu ◽  
Debra Piktel ◽  
Karen H. Martin ◽  
Werner J. Geldenhuys ◽  
...  

AbstractB-cell acute lymphoblastic leukemia (ALL) is characterized by accumulation of immature hematopoietic cells in the bone marrow, a well-established sanctuary site for leukemic cell survival during treatment. While standard of care treatment results in remission in most patients, a small population of patients will relapse, due to the presence of minimal residual disease (MRD) consisting of dormant, chemotherapy-resistant tumor cells. To interrogate this clinically relevant population of treatment refractory cells, we developed an in vitro cell model in which human ALL cells are grown in co-culture with human derived bone marrow stromal cells or osteoblasts. Within this co-culture, tumor cells are found in suspension, lightly attached to the top of the adherent cells, or buried under the adherent cells in a population that is phase dim (PD) by light microscopy. PD cells are dormant and chemotherapy-resistant, consistent with the population of cells that underlies MRD. In the current study, we characterized the transcriptional signature of PD cells by RNA-Seq, and these data were compared to a published expression data set derived from human MRD B-cell ALL patients. Our comparative analyses revealed that the PD cell population is markedly similar to the MRD expression patterns from the primary cells isolated from patients. We further identified genes and key signaling pathways that are common between the PD tumor cells from co-culture and patient derived MRD cells as potential therapeutic targets for future studies.


Blood ◽  
1995 ◽  
Vol 86 (2) ◽  
pp. 692-702 ◽  
Author(s):  
EJ Steenbergen ◽  
OJ Verhagen ◽  
EF van Leeuwen ◽  
H van den Berg ◽  
AE von dem Borne ◽  
...  

Crosslineage T-cell receptor delta (TCR delta) rearrangements are widely used as tumor markers for the follow up of minimal residual disease in childhood B-precursor acute lymphoblastic leukemia (ALL) by polymerase chain reaction (PCR). The major drawback of this approach is the risk of false-negative results due to clonal evolution. We investigated the stability of V delta 2D delta 3 rearrangements in a group of 56 childhood B-precursor ALL patients by PCR and Southern blot analysis. At the PCR level, V delta 2D delta 3-to-J alpha rearranged subclones (one pathway for secondary TCR delta recombination) were demonstrated in 85.2% of V delta 2D delta 3-positive patients tested, which showed that small subclones are present in the large majority of patients despite apparently monoclonal TCR delta Southern blot patterns. Sequence analysis of V delta 2D delta 3J alpha rearrangements showed a biased J alpha gene usage, with HAPO5 and J alpha F in 26 of 32 and 6 of 32 clones, respectively. Comparison of V delta 2D delta 3 rearrangement status between diagnosis and first relapse showed differences in seven of eight patients studied. In contrast, from first relapse onward, no clonal changes were observed in six patients studied. To investigate the occurrence of crosslineage TCR delta rearrangements in normal B and T cells, fluorescence-activated cell sorter-sorted peripheral blood CD19+/CD3- and CD19-/CD3+ cell populations from three healthy donors were analyzed. V delta 2D delta 3 rearrangements were detected at low frequencies in both B and T cells, which suggests that V delta 2-to-D delta 3 joining also occurs during normal B-cell differentiation. A model for crosslineage TCR delta rearrangements in B-precursor ALL is deduced that explains the observed clonal changes between diagnosis and relapse and is compatible with multistep leukemogenesis of B-precursor ALL.


Blood ◽  
1994 ◽  
Vol 83 (7) ◽  
pp. 1892-1902 ◽  
Author(s):  
H Cave ◽  
C Guidal ◽  
P Rohrlich ◽  
MH Delfau ◽  
A Broyart ◽  
...  

Abstract We have developed a strategy based on polymerase chain reaction (PCR) for detecting all possible gamma T-cell receptor (gamma TCR) rearrangements and the most common delta TCR rearrangements found in B- lineage and T-acute lymphoblastic leukemia (T-ALL). The segments amplified from blasts are then directly sequenced to derive clonospecific probes. From a series of 45 patients aged 1 to 15 years (42 B-lineage ALL, 3 T-ALL), 35 (83%) could be followed for minimal residual disease with at least one clonospecific probe. Detection of clonal markers using clonospecific probes routinely allowed the detection of 1 to 10 blasts out of 10(5) cells as determined by serial dilutions of the initial samples. Residual disease was quantitated by a competitive PCR assay based on the coamplification of an internal standard. Twenty children were prospectively followed for periods varying from 7 to 30 months. In most children, a progressive decrease of the tumor load was observed, and blasts became undetectable within 6 months after the initiation of treatment. A slower kinetics of decrease in tumor cells was found in three children. These three patients relapsed with blasts that continued to display the initial clonospecific markers. Three other children had a central nervous system relapse despite the absence of detectable medullary residual disease. The use of both delta and gamma TCR genes as clonal markers, as well as simplification in the methods to detect and quantify residual blasts reported here, will allow the study of the large number of patients required to determine the role of the detection of minimal residual disease by PCR in the follow-up of childhood ALL.


2015 ◽  
Vol 7 ◽  
pp. e2015001 ◽  
Author(s):  
Elena Maino ◽  
Anna Maria Scattolin ◽  
Piera Viero ◽  
Rosaria Sancetta ◽  
Anna Pascarella ◽  
...  

The introduction of newer cytotoxic monoclonal antibodies and the chimeric antigen receptor-modified T-cells is opening a new age in the management of B-lineage adult acute lymphoblastic leukemia. This therapeutic change must be very positively acknowledged because of the limits of intensive chemotherapy programs and allogeneic stem cell transplantation. With these traditional therapeutic tools in fact cure can be achieve in only 40-50% of the patients, with higher failure rates in the elderly, in those with post-induction persistence of minimal residual disease and especially in refractory/relapsed disease. The place of the novel immunotherapeutics in improving the outcome of adult patients with B-lineage acute lymphoblastic leukemia is reviewed.


Blood ◽  
1994 ◽  
Vol 83 (7) ◽  
pp. 1892-1902 ◽  
Author(s):  
H Cave ◽  
C Guidal ◽  
P Rohrlich ◽  
MH Delfau ◽  
A Broyart ◽  
...  

We have developed a strategy based on polymerase chain reaction (PCR) for detecting all possible gamma T-cell receptor (gamma TCR) rearrangements and the most common delta TCR rearrangements found in B- lineage and T-acute lymphoblastic leukemia (T-ALL). The segments amplified from blasts are then directly sequenced to derive clonospecific probes. From a series of 45 patients aged 1 to 15 years (42 B-lineage ALL, 3 T-ALL), 35 (83%) could be followed for minimal residual disease with at least one clonospecific probe. Detection of clonal markers using clonospecific probes routinely allowed the detection of 1 to 10 blasts out of 10(5) cells as determined by serial dilutions of the initial samples. Residual disease was quantitated by a competitive PCR assay based on the coamplification of an internal standard. Twenty children were prospectively followed for periods varying from 7 to 30 months. In most children, a progressive decrease of the tumor load was observed, and blasts became undetectable within 6 months after the initiation of treatment. A slower kinetics of decrease in tumor cells was found in three children. These three patients relapsed with blasts that continued to display the initial clonospecific markers. Three other children had a central nervous system relapse despite the absence of detectable medullary residual disease. The use of both delta and gamma TCR genes as clonal markers, as well as simplification in the methods to detect and quantify residual blasts reported here, will allow the study of the large number of patients required to determine the role of the detection of minimal residual disease by PCR in the follow-up of childhood ALL.


Sign in / Sign up

Export Citation Format

Share Document