Abstract 4575: Correlates of clinical response following autologous stem cell transplant and adoptive immunotherapy with engineered T cells expressing an affinity-enhanced T cell receptor in patients with mutliple myeloma.

Author(s):  
Aaron P. Rapoport ◽  
Edward A. Stadtmauer ◽  
Dan T. Vogl ◽  
Brendan Weiss ◽  
Gwendolyn K. Binder-Scholl ◽  
...  
Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 845-845
Author(s):  
Aaron P. Rapoport ◽  
Edward A Stadtmauer ◽  
Karen Chagin ◽  
Thomas Faitg ◽  
Malini Iyengar ◽  
...  

Abstract Background: NY-ESO-1 and LAGE-1a are cancer-testis antigens that are overexpressed in patients with multiple myeloma (MM), and the incidence of these antigens correlates with tumor proliferation and other high-risk features. Genetically engineered NY-ESO-1 SPEAR (specific peptide enhanced affinity receptor) T-cells (NY-ESO-1c259T cells) recognize the peptide sequence SLLMWITQC expressed by NY-ESO-1 or LAGE-1a in the context of HLA-A*02 presentation. This study evaluated treatment with NY-ESO-1SPEAR T-cells post-autologous stem cell transplant (ASCT) in patients with advanced MM. Methods: Eligible patients were HLA-A*02:01, 02:05 or 02:06 positive, with refractory, relapsed or high risk MM associated with one or more adverse cytogenetic abnormalities. Eligible patients' tumors also expressed NY-ESO-1 and/or LAGE-1a by qPCR. The primary study endpoint was safety. Secondary objectives included overall response rate (ORR) (sCR+CR+VGPR+PR) evaluated with the International Myeloma Working Group Criteria (Rajkumar S.V. et al., Blood 2011), progression-free survival (PFS), overall survival (OS), best overall response (BOR), duration of response (DOR), as well as gene-marked cell persistence. Lymphocytes were obtained by leukapheresis, isolated, activated, transduced to express NY-ESO-1c259T cell receptor, and expanded using anti-CD3/anti-CD28 immunomagnetic beads. While the SPEAR T-cells were being manufactured, stem cell mobilization was conducted using 1.5 g/m2 of cyclophosphamide plus G-CSF, and stem cells were collected (minimum: 2 × 106 CD34+ progenitors/kg). Once the manufactured product was ready, high-dose melphalan (140-200 mg/m2) was given 2 days before stem cell infusion. Two days after the stem cell infusion, the SPEAR T-cells were infused (median dose 3.1 × 109 of transduced T-cells, range 0.5-5.1 × 109). Disease was assessed at days 42, 100, 180, 270 and 360 post-T-cell infusion, and then every 3 months. Patients meeting the criteria for lenalidomide maintenance therapy received 10 mg/day starting around day 100 post-ASCT. Results: Twenty-five patients were enrolled, and all have been treated. Median age at enrollment was 61 yr (range 45 - 72); 60% were male. Based on analyses through July 2017, ORR at day 100 was 76% (1 sCR; 12 VGPR; 6PR), and at year 1, 13 patients were progression free (52%) of which 11 were responders (1 sCR; 1 CR; 8 VGPR; 1 PR). Three patients remain disease progression-free at 39, 56 and 61 months post T-cell infusion. Median PFS was ~13 months (range 3-61 months). Eleven of 25 patients (44%) are alive, and median survival was ~35 months (range 6-68 months). The most common adverse events (experienced by >70%) were diarrhea (100%), nausea (100%), anemia (96%), decreased appetite (92%), thrombocytopenia (92%), fatigue (88%), pyrexia (84%), rash (84%), hypokalemia (76%), febrile neutropenia (72%) and vomiting (72%). Autologous GVHD (24%) was reported in 6 patients (3 G3, 3 ≤G2); all resolved with corticosteroids and supportive therapy. No fatal adverse events have been reported. Conclusions: NY-ESO-1 SPEAR T-cell therapy in the setting of autologous stem cell transplant has promising efficacy and acceptable safety. GVHD, which manifests in a similar way as reported in prior transplant studies and is more frequent with adoptive T cell transfer (engineered or not engineered), appears manageable with appropriate supportive care. Analyses of transduced cell persistence, T-cell clonality, and minimal residual disease (MRD) genetic studies are ongoing and will be presented along with the efficacy and safety data for all 25 patients. Disclosures Chagin: Adaptimmune: Employment. Faitg: Adaptimmune: Employment. Iyengar: Adaptimmune: Employment. Trivedi: Adaptimmune: Employment. Norry: Adaptimmune: Employment. Holdich: Adaptimmune: Employment. Binder-Scholl: Adaptimmune: Employment. Amado: Adaptimmune: Employment. Fang: Adaptimmune: Employment.


2019 ◽  
Vol 3 (13) ◽  
pp. 2022-2034 ◽  
Author(s):  
Edward A. Stadtmauer ◽  
Thomas H. Faitg ◽  
Daniel E. Lowther ◽  
Ashraf Z. Badros ◽  
Karen Chagin ◽  
...  

Abstract This study in patients with relapsed, refractory, or high-risk multiple myeloma (MM) evaluated the safety and activity of autologous T cells engineered to express an affinity-enhanced T-cell receptor (TCR) that recognizes a peptide shared by cancer antigens New York esophageal squamous cell carcinoma-1 (NY-ESO-1) and L-antigen family member 1 (LAGE-1) and presented by HLA-A*02:01. T cells collected from 25 HLA-A*02:01-positive patients with MM expressing NY-ESO-1 and/or LAGE-1 were activated, transduced with self-inactivating lentiviral vector encoding the NY-ESO-1c259TCR, and expanded in culture. After myeloablation and autologous stem cell transplant (ASCT), all 25 patients received an infusion of up to 1 × 1010 NY-ESO-1 specific peptide enhanced affinity receptor (SPEAR) T cells. Objective response rate (International Myeloma Working Group consensus criteria) was 80% at day 42 (95% confidence interval [CI], 0.59-0.93), 76% at day 100 (95% CI, 0.55-0.91), and 44% at 1 year (95% CI, 0.24-0.65). At year 1, 13/25 patients were disease progression-free (52%); 11 were responders (1 stringent complete response, 1 complete response, 8 very good partial response, 1 partial response). Three patients remained disease progression-free at 38.6, 59.2, and 60.6 months post-NY-ESO-1 SPEAR T-cell infusion. Median progression-free survival was 13.5 months (range, 3.2-60.6 months); median overall survival was 35.1 months (range, 6.4-66.7 months). Infusions were well tolerated; cytokine release syndrome was not reported. No fatal serious adverse events occurred during study conduct. NY-ESO-1 SPEAR T cells expanded in vivo, trafficked to bone marrow, demonstrated persistence, and exhibited tumor antigen-directed functionality. In this MM patient population, NY-ESO-1 SPEAR T-cell therapy in the context of ASCT was associated with antitumor activity. This trial was registered at www.clinicaltrials.gov as #NCT01352286.


Oncotarget ◽  
2018 ◽  
Vol 9 (13) ◽  
pp. 11009-11019 ◽  
Author(s):  
Taigo Kato ◽  
Tatsuo Matsuda ◽  
Yuji Ikeda ◽  
Jae-Hyun Park ◽  
Matthias Leisegang ◽  
...  

Haematologica ◽  
2018 ◽  
Vol 104 (3) ◽  
pp. 622-631 ◽  
Author(s):  
Cornelia S. Link-Rachner ◽  
Anne Eugster ◽  
Elke Rücker-Braun ◽  
Falk Heidenreich ◽  
Uta Oelschlägel ◽  
...  

Blood ◽  
2005 ◽  
Vol 105 (2) ◽  
pp. 886-893 ◽  
Author(s):  
Xiaohua Chen ◽  
Raymond Barfield ◽  
Ely Benaim ◽  
Wing Leung ◽  
James Knowles ◽  
...  

Abstract The extent and rapidity with which T cells are regenerated from graft-derived precursor cells directly influences the incidence of infection and the T-cell–based graft-versus-tumor effect. Measurement of T-cell receptor excision circles (TRECs) in peripheral blood is a means of quantifying recent thymic T-cell production and has been used after transplantation in many studies to estimate thymus-dependent T-cell reconstitution. We hypothesized that the quality of thymic function before transplantation affects thymus-dependent T-cell reconstitution after transplantation. We used real-time polymerase chain reaction (PCR) to quantify signal-joint TRECs (sjTRECs) before and after transplantation. T-cell reconstitution was evaluated by T-cell receptor β (TCRβ) CDR3 size spectratyping. We tested 77 healthy sibling donors and 244 samples from 26 pediatric recipients of allogeneic hematopoietic stem cell transplantation (AHSCT). Blood from the healthy donors contained 1200 to 155 000 sjTREC copies/mL blood. Patients who had greater than 1200 copies/mL blood before transplantation showed early recovery of sjTREC numbers and TCRβ repertoire diversity. In contrast, patients who had fewer than 1200 copies/mL blood before transplantation demonstrated significantly slower restoration of thymus-dependent T cells. We conclude that the rate of reconstitution of thymus-dependent T cells is dependent on the competence of thymic function in the recipients before transplantation. Therefore, pretransplantation measurement of sjTREC may provide an important tool for predicting thymus-dependent T-cell reconstitution after transplantation.


2017 ◽  
Author(s):  
Tatsuo Matsuda ◽  
Taigo Kato ◽  
Yuji Ikeda ◽  
Matthias Leisegang ◽  
Sachiko Yoshimura ◽  
...  

2021 ◽  
Vol 7 (1) ◽  
pp. 48-56
Author(s):  
Matyas Ecsedi ◽  
Megan S. McAfee ◽  
Aude G. Chapuis

Sign in / Sign up

Export Citation Format

Share Document