scholarly journals Long-Term Outcomes of Patients with Epstein-Barr Virus-Driven Post-Transplant Lymphoproliferative Disease Following Solid Organ Transplant or Allogeneic Hematopoietic Cell Transplant Treated with Tabelecleucel in a Multicenter Expanded Access Program Study

2020 ◽  
Vol 26 (3) ◽  
pp. S61-S62
Author(s):  
Susan E Prockop ◽  
Ran Reshef ◽  
Donald E Tsai ◽  
Nancy Bunin ◽  
Rolla Abu-Arja ◽  
...  
Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 887-887
Author(s):  
Susan Prockop ◽  
Laurence Gamelin ◽  
Rajani Dinavahi ◽  
Yan Sun ◽  
Weizhi Zhao ◽  
...  

Abstract Introduction: Tabelecleucel is an investigational, off-the-shelf, allogeneic Epstein-Barr virus (EBV)-specific T-cell immunotherapy being studied in patients (pts) with serious EBV-driven diseases, including post-transplant lymphoproliferative disease (EBV + PTLD). Initial management of EBV + PTLD following solid organ transplant (SOT) and allogenic hematopoietic cell transplant (HCT) may include reduction in immunosuppression and/or off-label rituximab ± chemotherapy (CT). In pts with EBV + PTLD following HCT, median overall survival (OS) was ~1.7 months after failure of rituximab (Socié EBMT 2020) and ~3 months following SOT in pts who relapsed with rituximab and who did not respond to or did not receive CT (Zimmerman EHA 2019). We have previously shown that pts with EBV + PTLD who responded to tabelecleucel had clinical benefit, including >80% estimated 2-year survival rates (Prockop JCI 2020, Prockop EBMT 2021, Prockop ATC 2021). Here, we report aggregate OS across pts with EBV + PTLD following HCT and SOT (combined population) by best overall response (BOR; eg, CR or PR) with tabelecleucel treatment. Methods: Treatment response and OS were assessed in three open-label studies (NCT00002663, NCT01498484 and NCT02822495). Analyses based on pooled data from the three studies were performed. All pts received tabelecleucel at ~2 x 10 6 cells/kg on Days 1, 8 and 15 in 35-day treatment cycles. Pts who did not initially respond could switch to tabelecleucel with a different human leukocyte antigen (HLA) restriction (restriction switch), and were treated until unacceptable toxicity, maximal response, or up to four different HLA restrictions. The length of post-treatment follow-up (if any) varied per protocol. Results: In this combined analysis, 76 pts with EBV + PTLD relapsed or refractory to rituximab (HCT; n=50) or rituximab ± CT (SOT; n=26) were treated with a median (range) of 2 (1-9) cycles of tabelecleucel. The investigator-assessed objective response rate, defined as the proportion of pts with BOR of CR or PR, was 63.2% overall (48/76), including 32 pts with CR and 16 pts with PR (Table 1, Figure 1). The median OS (95% confidence interval [CI]) for all treated pts was 54.6 months (14.8, 115.0) after a median follow-up (range) of 14.8 (0.4-115.0) months. The estimated overall 1- and 2-year OS rates (95% CI) were 65.8% (53.6, 75.5) and 57.8% (45.4, 68.5), respectively. The estimated 1- and 2-year OS rates (95% CI) in responders (CR+PR, n=48) were 91.3% (78.4, 96.6) and 86.2% (71.7, 93.6), respectively. Two-year estimated OS rates (95% CI) were 86.2% (67.0, 94.6) and 86.5% (55.8, 96.5) for pts with CR and PR, respectively (Table 1). Treatment was well tolerated with no confirmed evidence for graft vs host disease, cytokine release syndrome, SOT rejection, or neurologic events attributable to tabelecleucel in these very sick, treatment refractory, and immunocompromised pts. Conclusions: Tabelecleucel was well tolerated. Additionally, the combined data reported here show that pts whose EBV + PTLD failed to respond to rituximab (HCT) or rituximab ± CT (SOT) and who responded to tabelecleucel experienced long-term survival. Importantly, pts who achieved a PR with tabelecleucel derived similar OS benefit to those who achieved a CR. These results are consistent with those reported previously in separate subsets of pts with EBV + PTLD following HCT (Prockop EBMT 2021) or SOT (Prockop ATC 2021). Figure 1 Figure 1. Disclosures Prockop: Memorial Sloan Kettering Cancer Center: Other: S Prockop receives support for the conduct of sponsored clinical trials through MSK from Atara Biotherapeutics, Jasper and AlloVir. , Patents & Royalties: S Prockop is a co-inventor on intellectual property (IP) licensed to Atara. S Prockop has waived rights to this IP to MSK and has no personal financial interests in Atara. MSK has financial interests in Atara and IP interests relevant to this abstract. ; Atara Biotherapeutics: Other: support for the conduct of sponsored trials and Inventor; Jasper: Other: support for the conduct of sponsored trials; AlloVir: Other: support for the conduct of sponsored trials; Neovii: Consultancy; ADMA Biologics: Consultancy; MSK: Other: Inventor. Gamelin: Atara Biotherapeutics: Current Employment. Dinavahi: Atara Biotherapeutics: Current Employment. Sun: Atara Biotherapeutics: Current Employment. Zhao: Atara Biotherapeutics: Current Employment. Galderisi: Atara Biotherapeutics: Current Employment. Mehta: Atara Biotherapeutics: Current Employment.


2014 ◽  
Vol 155 (8) ◽  
pp. 313-318
Author(s):  
Anita Stréhn ◽  
László Szőnyi ◽  
Gergely Kriván ◽  
Lajos Kovács ◽  
György Reusz ◽  
...  

Introduction: Among possible complications of transplantation the post-transplant lymphoproliferative disease due to immunosuppressive therapy is of paramount importance. In most cases the direct modulating effect of Epstein–Barr virus on immune cells can be documented. Aim: The aim of the authors was to evaluate the incidence os post-transplant lymphoproliferative diseases in pediatric transplant patients in Hungary. Method: The study group included kidney, liver and lung transplant children followed up at the 1st Department of Pediatrics, Semmelweis University, Budapest and stem cell transplant children at Szent László Hospital, Budapest. Data were collected from 78 kidney, 109 liver and 17 lung transplant children as well as from 243 children who underwent allogenic stem cell transplantation. Results: Between 1998 and 2012, 13 children developed post-transplant lymphoproliferative disorder (8 solid organ transplanted and 5 stem cell transplanted children). The diagnosis was based on histological findings in all cases. Mortality was 3 out of the 8 solid organ transplant children and 4 out of the 5 stem cell transplant children. The highest incidence was observed among lung transplant children (17.6%). Conclusions: These data indicate that post-transplant lymphoproliferative disease is a rare but devastating complication of transplantation in children. The most important therapeutic approaches are reduction of immunosuppressive therapy, chemotherapy and rituximab. Early diagnosis may improve clinical outcome and, therefore, routine polymerase chain reaction screening for Epstein–Barr virus of high risk patients is recommended. Orv. Hetil., 2014, 155(8), 313–318.


Blood ◽  
1994 ◽  
Vol 84 (3) ◽  
pp. 972-984 ◽  
Author(s):  
SA Riddler ◽  
MC Breinig ◽  
JL McKnight

Epstein-Barr virus (EBV)-associated posttransplant lymphoproliferative disease (PTLD) is an uncommon but potentially fatal complication of immunosuppression in solid-organ transplant recipients. A semiquantitative DNA polymerase chain reaction assay was developed to amplify a unique 269-bp region of the EBNA-1 gene in peripheral blood lymphocytes (PBL) using the primers described by Telenti et al (J Clin Microbiol 28:2187, 1990). Serial samples were studied from 23 transplant recipients, 12 of whom were diagnosed with PTLD. The majority of transplant recipients who were EBV seropositive at the time of transplant surgery and who did not develop PTLD (5 of 7, 71%) exhibited less than a 10-fold increase in the levels of EBV-infected PBL over the 0.1 to 5 EBV genomes/10(6) PBL observed in immunocompetent EBV seropositive controls. Transplant recipients who were seronegative at the time of transplantation and who underwent a primary EBV infection but did not develop PTLD exhibited a reduced capacity to control viremia because the levels of EBV-infected PBL were up to 400 times greater than the 1.0 to 50 EBV genomes/10(6) PBL observed in individuals undergoing acute infectious mononucleosis (Rocci et al: N Engl J Med 296:132, 1977). However, all transplant recipients who developed PTLD exhibited a marked elevation of EBV-infected PBL independent of their serologic state at the time of transplantation. Six of the 10 transplant recipients with PTLD exhibited > or = 300,000 EBV genomes/10(5) PBL, two exhibited 10,000 to 50,000 EBV-infected genomes/10(5) PBL, and one each exhibited 2,500 and 500 EBV genomes/10(5) PBL. However, the latter two samples were obtained 4 to 5 weeks after the diagnosis of PTLD and may reflect a decrease in viral load resulting from immunomodulation. Marked decreases in the levels of EBV nuclear antigen-1 (EBNA-1), EBNA-2, and EBNA-LP antibodies correlated with the increase in EBV-infected PBL. Hence, a quantitative difference in circulating EBV viral load and EBNA antibody levels is evident between transplant recipients with and without PTLD and may be useful as a noninvasive prognostic marker with which to monitor and/or predict the development of PTLD.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 301-301
Author(s):  
Susan Prockop ◽  
Kris Michael Mahadeo ◽  
Amer Beitinjaneh ◽  
Sylvain Choquet ◽  
Patrick Stiff ◽  
...  

Abstract Background: Tabelecleucel is an investigational, off-the-shelf, allogeneic Epstein-Barr virus (EBV)-specific T-cell immunotherapy being studied in patients (pts) with serious EBV-driven diseases, including post-transplant lymphoproliferative disease (EBV + PTLD). The median overall survival (OS) after treatment failure of rituximab is short at ~1.7 months in EBV + PTLD following hematopoietic cell transplant (HCT) (Socié EBMT 2020) and ~3 months in solid organ transplant (SOT) recipients with EBV + PTLD whose disease relapsed after rituximab and who did not respond to or did not receive chemotherapy (CT) (Zimmerman EHA 2019). These poor outcomes demonstrate an urgent unmet need for effective therapies in this ultra-rare serious disease. Promising outcomes have previously been demonstrated (Prockop JCI 2020). Additionally, we have shown clinical benefit with a >60% objective response rate (ORR) and >80% estimated 2-year OS rates (Prockop EBMT 2021, Prockop ATC 2021) in pts with EBV + PTLD, irrespective of pts achieving a complete response (CR) or partial response (PR) to tabelecleucel. Safety data have been assessed in >150 pts with EBV + PTLD treated with tabelecleucel with tumor flare reaction (TFR) as the only identified risk. Here, for the first time, we report interim efficacy and safety results from the pivotal phase 3 clinical trial ALLELE (NCT03394365) - a multicenter, open-label study of tabelecleucel after failure of rituximab ± CT in pts with EBV + PTLD following SOT or HCT. Methods: ALLELE is evaluating the clinical benefit of tabelecleucel in two cohorts: (1) pts with EBV + PTLD following SOT after failure of rituximab ± CT (n=33), and (2) pts with EBV + PTLD following HCT after failure of rituximab monotherapy (n=33). Tabelecleucel is administered at a dose of 2 x 10 6 cells/kg on days 1, 8, and 15, followed by observation through each cycle lasting 35 days. Response per clinical and radiographic assessment (by PET/CT) is evaluated by the investigator and by independent review using Lugano Classification with LYRIC modification. Key efficacy endpoints include ORR, duration of response (DOR), time to response (TTR), and OS. After treatment is completed or discontinued, pts are assessed every 3 months up to 24 months, and every 6 months thereafter for survival status up to 5 years. Results: As of May 2021, 38 pts (24 SOT, 14 HCT) were evaluable for response assessment by independent oncologic response adjudication (IORA) and had the opportunity for 6 months follow-up (Table 1). SOT and HCT pts received a median (range) of 2.0 (1-6) and 3.0 (1-5) cycles of tabelecleucel, respectively. ORR was 50% (19/38, 95% CI: 33.4, 66.6) in the overall population, 50.0% (12/24, 95% CI: 29.1, 70.9) in SOT, and 50.0% (7/14, 95% CI: 23.0, 77.0) in HCT, with a best overall response of CR (n=5, SOT; n=5, HCT) or PR (n=7, SOT; n=2, HCT; Table 2). Overall, median TTR was 1.1 months (0.7-4.7), 11 of 19 responders had a DOR lasting >6 months, and median DOR was not reached (Table 2). Median OS was 18.4 (95% CI: 6.9, NE) months overall, 16.4 (95% CI: 3.5, NE) months for SOT, and not yet reached for HCT. 1-year survival rate was 61.1% (95% CI: 42.9, 75.0) overall, 57.4% (95% CI: 35.2, 74.5) for SOT, and 66.8% (95% CI: 32.4, 86.6) for HCT. Those who responded had a longer survival compared to the non-responders, with a median OS of NE (95% CI: 16.4, NE) and 1-year survival rate of 89.2% (95% CI: 63.1, 97.2). Non-responders had a median OS of 5.7 (95% CI: 1.8, 12.1) months and 1-year survival rate of 32.4% (95% CI: 12.1, 54.9) (Table 3). Serious treatment emergent AEs (TEAEs) were reported in 62.5% of SOT and 57.1% of HCT pts. Fatal TEAEs were reported in 16.7% of SOT and 7.1% of HCT pts; none of the fatal TEAEs were related to study treatment. Tabelecleucel was well-tolerated with no reports of TFR and no confirmed evidence for graft vs host disease, organ rejection, infusion reactions, or cytokine release syndrome in relation to tabelecleucel in these treatment refractory and immunocompromised pts. Conclusions: Tabelecleucel phase 3 interim data are reported here for the first time and show clinically meaningful outcomes and promising ORR and OS in a pt population with no approved treatment options and otherwise poor survival. Tabelecleucel, an allogeneic cell therapy, was well tolerated without evidence of safety concerns typically observed with autologous chimeric antigen receptor cell therapies. Figure 1 Figure 1. Disclosures Prockop: Memorial Sloan Kettering Cancer Center: Other: S Prockop receives support for the conduct of sponsored clinical trials through MSK from Atara Biotherapeutics, Jasper and AlloVir. , Patents & Royalties: S Prockop is a co-inventor on intellectual property (IP) licensed to Atara. S Prockop has waived rights to this IP to MSK and has no personal financial interests in Atara. MSK has financial interests in Atara and IP interests relevant to this abstract. ; Atara Biotherapeutics: Other: support for the conduct of sponsored trials and Inventor; Jasper: Other: support for the conduct of sponsored trials; AlloVir: Other: support for the conduct of sponsored trials; Neovii: Consultancy; ADMA Biologics: Consultancy; MSK: Other: Inventor. Mahadeo: Atara Biotherapeutics: Consultancy. Beitinjaneh: Kite/Gilead: Other: Ad Board Event Attendee. Choquet: Sanofi, Celegene, Roche, Abbvie, Sandoz, Janssen, Takeda: Consultancy. Stiff: Cellectar: Research Funding; CRISPR: Consultancy; Gamida-Cell, Atara, Amgen, Incyte, Takeda, Macrogenetics, Eisai: Research Funding. Reshef: Bayer, BMS, Regeneron, TScan, Synthekine, Atara, Jasper: Consultancy. Dahiya: Kite, a Gilead Company: Consultancy; Miltenyi Biotech: Research Funding; Atara Biotherapeutics: Consultancy; Jazz Pharmaceuticals: Research Funding; BMS: Consultancy. Parmar: Atara Biotherapeutics: Current Employment. Ye: Atara Biotherapeutics: Current Employment. Gamelin: Atara Biotherapeutics: Current Employment. Dinavahi: Atara Biotherapeutics: Current Employment.


Blood ◽  
1994 ◽  
Vol 84 (3) ◽  
pp. 972-984 ◽  
Author(s):  
SA Riddler ◽  
MC Breinig ◽  
JL McKnight

Abstract Epstein-Barr virus (EBV)-associated posttransplant lymphoproliferative disease (PTLD) is an uncommon but potentially fatal complication of immunosuppression in solid-organ transplant recipients. A semiquantitative DNA polymerase chain reaction assay was developed to amplify a unique 269-bp region of the EBNA-1 gene in peripheral blood lymphocytes (PBL) using the primers described by Telenti et al (J Clin Microbiol 28:2187, 1990). Serial samples were studied from 23 transplant recipients, 12 of whom were diagnosed with PTLD. The majority of transplant recipients who were EBV seropositive at the time of transplant surgery and who did not develop PTLD (5 of 7, 71%) exhibited less than a 10-fold increase in the levels of EBV-infected PBL over the 0.1 to 5 EBV genomes/10(6) PBL observed in immunocompetent EBV seropositive controls. Transplant recipients who were seronegative at the time of transplantation and who underwent a primary EBV infection but did not develop PTLD exhibited a reduced capacity to control viremia because the levels of EBV-infected PBL were up to 400 times greater than the 1.0 to 50 EBV genomes/10(6) PBL observed in individuals undergoing acute infectious mononucleosis (Rocci et al: N Engl J Med 296:132, 1977). However, all transplant recipients who developed PTLD exhibited a marked elevation of EBV-infected PBL independent of their serologic state at the time of transplantation. Six of the 10 transplant recipients with PTLD exhibited > or = 300,000 EBV genomes/10(5) PBL, two exhibited 10,000 to 50,000 EBV-infected genomes/10(5) PBL, and one each exhibited 2,500 and 500 EBV genomes/10(5) PBL. However, the latter two samples were obtained 4 to 5 weeks after the diagnosis of PTLD and may reflect a decrease in viral load resulting from immunomodulation. Marked decreases in the levels of EBV nuclear antigen-1 (EBNA-1), EBNA-2, and EBNA-LP antibodies correlated with the increase in EBV-infected PBL. Hence, a quantitative difference in circulating EBV viral load and EBNA antibody levels is evident between transplant recipients with and without PTLD and may be useful as a noninvasive prognostic marker with which to monitor and/or predict the development of PTLD.


Sign in / Sign up

Export Citation Format

Share Document