scholarly journals Trial-in-Progress: Brentuximab Vedotin in Combination with Lenalidomide and Rituximab in Subjects with Relapsed or Refractory Diffuse Large B-Cell Lymphoma (DLBCL)

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 14-14
Author(s):  
Nancy L. Bartlett ◽  
Christopher A. Yasenchak ◽  
Robert B. Sims ◽  
Grzegorz S. Nowakowski

Background Majority of patients with relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL) who relapse after HSCT, or who are not candidates for HSCT have poor outcomes and are in need of novel therapies. Preclinical data provides a strong rationale for combining brentuximab vedotin, lenalidomide, and rituximab in the treatment of R/R DLBCL. In a phase 1 trial, 37 subjects with R/R DLBCL were treated with brentuximab vedotin in combination with lenalidomide. The objective response rate (ORR) (proportion of subjects with complete response [CR] or partial response [PR]) for all subjects was 56.7% (21 of 37 subjects). For the CD30-positive population, the ORR was 73.3% (11 of 15 subjects), whereas the ORR for the CD30 <1% population was 45.6% (10/22). The median follow-up time was 14.3 months (range: 1 to 56.7 months). The median duration of remission for subjects with CR or PR was 13.2 months (range: 3.2 to 55.2 months). For subjects with CR, PR, or stable disease (SD) >6 months, the median duration of remission was 11.70 months (range: 3.2 to 55.2 months). The progression-free survival (PFS) and overall survival (OS) results appear similar in both the CD30-positive and CD30 <1% groups. The PFS in the combined population is 10.2 months and the median OS is 14.2 months (manuscript in preparation). Study Design and Methods This is a randomized, double-blind, placebo-controlled, active-comparator, multicenter phase 3 study designed to evaluate the efficacy of brentuximab vedotin in combination with lenalidomide and rituximab versus placebo in combination with lenalidomide and rituximab for the treatment of subjects with R/R DLBCL (NCT04404283). Key eligibility criteria include the following: subjects aged 18 and older with R/R DLBCL with an eligible subtype; subjects must have ≥2 prior lines of therapy and must be ineligible for, or have declined, stem cell transplant, and chimeric antigen receptor T-cell (CAR-T) therapy; subjects must have an ECOG performance status score of 0 to 2; subjects must have a fluorodeoxyglucose-avid disease by PET and bidimensional measurable disease of at least 1.5 cm by CT, as assessed by the site radiologist. Subjects (n=400) will be randomized 1:1 to receive either brentuximab vedotin or placebo in combination with lenalidomide and rituximab and will be stratified by CD30 expression (positive [≥1%] versus <1%), prior allogeneic or autologous stem cell transplant therapy (received or not), prior CAR-T therapy (received or not), and cell of origin (germinal-center B-cell-like (GCB) or non-GCB). Cell of origin (GCB or non-GCB) will be histologically determined by local pathology assessment. Disease response will be assessed by a blinded independent central review (BICR) and the investigator according to the Lugano Classification Revised Staging System. Radiographic disease evaluations, including contrast-enhanced CT scans and PET/CT, will be assessed at baseline, then every 6 weeks from randomization for 12 months, then every 12 weeks (±3 days). PET/CTs will be discontinued if negative. For the primary efficacy analyses of PFS per BICR in the intent-to-treat population and in CD30-positive subjects, the stratified log-rank test will be used to compare PFS between the 2 treatment groups. The hazard ratio will be estimated using a stratified Cox regression model. PFS will also be summarized using the Kaplan-Meier method. Similar methods will be used for the secondary efficacy endpoint of OS, and other time-to-event efficacy endpoints. The trial will have sites open in the US and multiple countries in Europe and Asia, with enrollment planning to begin in the second half of 2020. Disclosures Bartlett: Roche/Genentech: Consultancy, Research Funding; Seattle Genetics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Millennium: Research Funding; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pharmacyclics: Research Funding; Seattle Genetics: Consultancy, Research Funding; Janssen: Research Funding; Kite, a Gilead Company: Research Funding; Merck: Research Funding; BTG: Consultancy; Acerta: Consultancy; Affimed Therapeutics: Research Funding; Immune Design: Research Funding; Forty Seven: Research Funding; BMS/Celgene: Research Funding; ADC Therapeutics: Consultancy; Autolus: Research Funding. Yasenchak:Takeda: Honoraria; BeiGene: Speakers Bureau; Seattle Genetics: Honoraria, Research Funding. Sims:Seattle Genetics, Inc.: Current Employment, Current equity holder in publicly-traded company, Other: Travel expenses. Nowakowski:Roche: Consultancy, Research Funding; Celgene/BMS: Consultancy, Research Funding; Kite: Consultancy; Denovo: Consultancy; Kymera: Consultancy; Ryvu: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other; Curis: Consultancy; Seattle Genetics: Consultancy; Nanostrings: Research Funding; MorphoSys: Consultancy, Research Funding.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 96-96 ◽  
Author(s):  
Dahlia Sano ◽  
Loretta J. Nastoupil ◽  
Nathan H. Fowler ◽  
Luis Fayad ◽  
F. B. Hagemeister ◽  
...  

Abstract Background Axicabtagene ciloleucel (axi-cel) is an autologous CD19-specific CAR T-cell therapy product that was FDA approved for the treatment of adult patients with relapsed or refractory large B-cell lymphoma after at least two lines of systemic therapy. In the pivotal ZUMA-1 study, the best overall response (ORR) and complete response (CR) rates observed in 108 patients treated with axi-cel were 82% and 58%, respectively. At a median follow-up of 15.4 months, 42% of the patients remain in ongoing response (Neelapu et al. N Eng J Med 2017). Analysis of efficacy outcomes in patients <65 years (N=81) and ³65 years (N=27) showed that the ORR and ongoing response at 12 months were comparable between the two subgroups (Neelapu et al. N Eng J Med 2017). Whether the safety is also comparable between the two subgroups is unknown. Here, we report safety outcomes in elderly patients (³65 years) with large B-cell lymphoma treated with axi-cel at our institution. Methods We retrospectively analyzed and reviewed the data from patients treated with axi-cel at our institution. Patients had a diagnosis of relapsed or refractory diffuse large B-cell lymphoma (DLBCL), primary mediastinal B-cell lymphoma (PMBCL), high-grade B-cell lymphoma (HGBCL), and transformed follicular lymphoma (TFL). Patients were treated with conditioning chemotherapy with cyclophosphamide and fludarabine for 3 days followed by axi-cel infusion after 2 days of rest at a dose of 2 x 106 CAR+ T cells/kg body weight. Patients were monitored for toxicities for at least 7 days in the hospital after CAR T infusion and those who had at least 30 days of follow-up after axi-cel were considered to be evaluable for safety. Cytokine release syndrome (CRS) and neurological toxicity termed as CAR-related encephalopathy syndrome (CRES) were graded according to the CARTOX grading system (Neelapu et al. Nat Rev Clin Oncol 2018). Results A total of 61 patients with relapsed or refractory large B-cell lymphoma who received axi-cel at our institution were included. Of these, 44 (72%) patients were <65 years of age and 17 (28%) patients were ³65 years of age. The baseline characteristics of the patients are summarized in Table 1. ORR and CR rates at Day 30 were comparable between the two groups. CRS was common in both groups and was observed in 83% and 91% of the patients in the older and younger age groups, respectively. But most CRS events were grade 1-2. Grade 3 or higher CRS was observed in 18% vs. 11% in the older vs. younger age groups (P=0.67). One patient with a history of autoimmune disease in the elderly group died of hemophagocytic lymphohistiocytosis (HLH). CRES was observed in 58% and 71% of the patients in the older and younger age groups, respectively. Grade 3 or higher CRES was observed in 29% vs. 39% in the older vs. younger age groups (P=0.58). Median hospitalization period for axi-cel CAR T-cell therapy was comparable between the two groups. Conclusions Our results suggest that response rates are comparable between the elderly and younger age groups at day 30 after axi-cel therapy. Importantly, toxicities due to CRS and/or CRES after axi-cel CD19 CAR T cell therapy are comparable between the elderly (³65 years) and younger (<65 years) patients with relapsed or refractory large B-cell lymphoma. Table 1 Table 1. Disclosures Nastoupil: Merck: Honoraria, Research Funding; Janssen: Research Funding; Juno: Honoraria; Novartis: Honoraria; Genentech: Honoraria, Research Funding; TG Therappeutics: Research Funding; Karus: Research Funding; Celgene: Honoraria, Research Funding; Spectrum: Honoraria; Gilead: Honoraria. Fowler:Pharmacyclics: Consultancy, Research Funding; Janssen: Consultancy, Research Funding. Samaniego:ADC Therapeutics: Research Funding. Wang:Kite Pharma: Research Funding; Acerta Pharma: Honoraria, Research Funding; Novartis: Research Funding; Juno: Research Funding; Pharmacyclics: Honoraria, Research Funding; Dava Oncology: Honoraria; AstraZeneca: Consultancy, Research Funding; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; MoreHealth: Consultancy; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Westin:Kite Pharma: Membership on an entity's Board of Directors or advisory committees; Apotex: Membership on an entity's Board of Directors or advisory committees; Novartis Pharmaceuticals Corporation: Membership on an entity's Board of Directors or advisory committees; Celgen: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4225-4225
Author(s):  
Oscar Calzada ◽  
Kyle T. Bradley ◽  
Jeffrey Switchenko ◽  
Ashley D. Staton ◽  
Jean L. Koff ◽  
...  

Abstract Introduction: Prior series have identified CD30 expression by immunohistochemistry (IHC) is associated with improved overall survival (OS) in patients with newly diagnosed diffuse large B-cell lymphoma (DLBCL; 5-year OS: 79% vs 59% in CD30(-) patients, Hu et al, Blood 2013). It is unclear, however, whether CD30(+) status is retained throughout the course of the disease. Conversely, it is also unknown whether patients with CD30(-) tumors at diagnosis may present with CD30(+) disease upon relapse. As there is a currently approved antibody-drug conjugate targeting CD30, brentuximab vedotin, an improved understanding of CD30 expression in DLBCL may inform therapy options for relapsed (and potentially newly diagnosed) patients. Here, we evaluated patients with relapsed DLBCL with available tissue samples, including those with paired tissue samples from the time of diagnosis to assess for CD30 status for the duration of their disease course. Methods: This cohort included patients ≥ 18 years old with relapsed DLBCL for whom biopsy samples and clinical data were available. Tissue samples at diagnosis and from time of relapse were collected from our institution's pathology archive, and IHC-staining for CD30 expression was performed on all available involved tissue. CD30 status was assessed using a comprehensive form including assessment of percentage of CD30(+) cells and distribution of staining within each cell. Both neoplastic and surrounding non-neoplastic cells were evaluated. All assessment of CD30 staining was completed by one hematopathologist. We also collected comprehensive clinical, demographic and pathologic data for each patient. Results: We identified 25 patients with relapsed/refractory DLBCL with available tissue samples from the time of relapse, including 12 patients with available paired diagnostic tissue. Among all patients, the median age at diagnosis was 58 years (range 34-76), 48% were male, 56% were stage III/IV, and 62% presented with B-symptoms. Eighty-eight percent of patients received R-CHOP as frontline therapy. Cell of origin by the Hans algorithm was germinal center B-cell-like (GCB) for 9 patients, non-GCB for 5 patients, and unknown for 11 patients. After pathologic review, all 25 samples were CD30-negative at relapse, including all 12 paired samples which were CD30-negative at diagnosis and relapse, suggesting that CD30 expression does not appear to be acquired at the time of relapse in DLBCL patients who present with CD30-negative disease. Conclusions: This retrospective, single-center cohort analysis suggests that patients with newly diagnosed DLBCL with tumors negative for CD30 expression retain CD30-negative status at relapse. Thus, assaying biopsies at relapse for CD30 positivity by IHC to investigate candidacy for salvage treatment with brentuximab vedotin in a patient with a tumor previously CD30(-) may be of limited value. Alternative computer-aided methods to assess CD30 expression in samples that are considered negative by conventional IHC may better identify the presence of CD30 among patients with relapsed DLBCL. Disclosures Calzada: Seattle Genetics: Research Funding. Flowers:Seattle Genetics: Research Funding; Optum Rx, Seattle Genetics, Genentech/Roche: Consultancy; Spectrum, Janssen, Infinity, AbbVie, Acerta, Pharmacyclics, TG Therapeutics: Research Funding; Celgene Corporation: Consultancy, Honoraria; Millennium: Consultancy, Research Funding; Gilead: Consultancy, Research Funding. Cohen:Seattle Genetics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Millennium/Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pharmacyclics: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Infinity: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bristol-Myers Squibb: Research Funding.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4395-4395 ◽  
Author(s):  
Bertrand Coiffier ◽  
Catherine Thieblemont ◽  
Sophie de Guibert ◽  
Jehan Dupuis ◽  
Vincent Ribrag ◽  
...  

Abstract Background SAR3419 is a humanized anti-CD19 antibody conjugated to maytansin DM4, a potent cytotoxic agent. SAR3419 targets CD19, an antigen expressed in the majority of B cell non-Hodgkin lymphomas (NHL). The recommended dose for single agent SAR3419 was previously determined to be 55 mg/m2 administered IV every week for 4 weeks, then bi-weekly. In phase I, clinical activity was shown mainly in patients with follicular lymphoma (FL) and diffuse large B-cell lymphoma (DLBCL). (Trial funded by Sanofi). Methods Patients (pts) with a CD20+ and CD19+ DLBCL relapsing or refractory (R/R) after at least 1 standard treatment including rituximab and not candidate for or who already underwent transplantation, were eligible. Refractory disease was defined as unresponsive to or progressing within 6 months of regimen completion. Fresh (or recent formalin-fixed, paraffin-embedded) biopsy was required before SAR3419 start. Pts received 375 mg/m2 of rituximab (R) IV and 55 mg/m² of SAR3419 on day 1, 8, 15, 22 (35-day cycle 1), followed by bi-weekly R and SAR3419 at the same doses for 2 additional 28-day cycles, provided there was no disease progression or other study discontinuation criteria met. The primary objective was the overall response rate (ORR) following Cheson 2007 criteria, with the first tumor assessment being done 42 days after the last study treatment administration. Secondary objectives were: safety, pharmacokinetics (PK), duration of response (DOR), progression free survival (PFS), overall survival (OS) and correlation of the antitumor and biological activity of the combination with tumor biomarker status. Results Fifty-three pts were enrolled, 52 treated. Median age was 66.5 years (range 38-85), 50% were male; 23%, 33% and 40% of patients had received 1, 2 or ≥3 prior chemo/immunotherapy regimens for DLBCL, respectively. Of the enrolled patients, 3.8% had received no prior regimen for DLBCL and therefore were excluded from primary analysis for efficacy. Seventy-three percent had stage III/IV disease, 59% had elevated lactate dehydrogenase (LDH), and 63% had bulky disease. Sixty percent were refractory to first regimen (primary refractory), 16% were refractory to last regimen and 24% were relapsed pts. The ORR in the per-protocol population (n=45) was 31.1% (80% confidence interval (CI): 22.0% to 41.6%). Among the 14 responders, 5 had progressed at the time of analysis, with duration of response beyond 6 months for 3 of them. The ORR was 58.3% (80% CI: 36.2% to 78.1%) for patients with relapsed DLBCL (n=12), 42.9% (80% CI: 17.0% to 72.1%) for pts refractory to last regimen (n=7) and 15.4% (80% CI: 6.9% to 28.4%) for primary refractory pts (n=26). Overall survival and PFS data are not yet mature. Biomarkers and PK data will be presented at the meeting. The most common (≥10%) all grades non-hematologic treatment-emergent adverse events (TEAEs) were asthenia (25.0%), nausea (21.2%), cough (19.2%), diarrhea (17.3%), weight decrease (17.3%), vomiting (15.4%), dyspnea (15.4%), abdominal pain (13.5%), back pain (13.5%), pyrexia (13.5%) and constipation (11.5%). Related grade 3-4 TEAEs were: 1 syncope, 1 bronchospasm, 2 neutropenia and 1 anemia. No TEAEs led to treatment discontinuation, no grade 3-4 peripheral neuropathy or grade 3-4 ocular events were observed. Two pts experienced grade 2 keratitis, both rapidly recovered with local treatment. Hematological toxicity was moderate, with grade 3-4 neutropenia and thrombocytopenia in 15.7% and 9.8% pts, respectively. No complications related to neutropenia were reported. Grade 3 transaminase increase was observed in 1 patient. Conclusions The combination of SAR3419 plus R showed moderate ORR in R/R DLBCL; however the study population was of poor prognosis (60% refractory to first line therapy). In the relapsed DLBCL patients a higher ORR was observed. SAR3419 plus R presented with a favorable safety profile. Further investigations on biomarker expression are ongoing to identify a sub-group of pts who could have better benefited from this combination. Disclosures: Coiffier: Sanofi: Membership on an entity’s Board of Directors or advisory committees. Off Label Use: Phase II of SAR3419. Ribrag:Johnson & Johnson: Honoraria, Membership on an entity’s Board of Directors or advisory committees; Sanofi: Consultancy, Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding; Bayer: Research Funding; Takeda: Membership on an entity’s Board of Directors or advisory committees; Servier: Membership on an entity’s Board of Directors or advisory committees, Research Funding. Cartron:LFB: Honoraria; GSK: Honoraria; Roche: Consultancy, Honoraria, Speakers Bureau. Casasnovas:Roche: Consultancy, Honoraria, Research Funding. Hatteville:Sanofi: Employment. Zilocchi:Sanofi: Employment. Oprea:Sanofi: Employment. Tilly:Amgen: Research Funding; Janssen: Honoraria; Pfizer: Honoraria; Takeda: Membership on an entity’s Board of Directors or advisory committees; Roche: Honoraria; Celgene: Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3564-3564
Author(s):  
Nancy L. Bartlett ◽  
Christopher A. Yasenchak ◽  
Khaleel Ashraf ◽  
William Harwin ◽  
Robert Sims ◽  
...  

Abstract Background The majority of patients with relapsed/refractory (R/R) DLBCL who relapse after hematopoietic stem cell transplantation (HSCT) or chimeric antigen receptor T-cell (CAR-T) therapy, or who are not candidates for HSCT or CAR-T, have poor outcomes and need novel therapies (Crump 2017). Brentuximab vedotin (BV) was the first antibody-drug conjugate to be approved in multiple cancer types (Gauzy-Lazo 2020). The combination of a CD30-directed monoclonal antibody, a protease-cleavable linker, and the microtubule-disrupting agent monomethyl auristatin E drives the anticancer activity of BV by inducing cell cycle arrest and apoptosis (Sutherland 2006). Lenalidomide may enhance the activity of BV through immune-mediated mechanisms. In a phase 1 trial (NCT02086604) in which 37 patients with R/R DLBCL were treated with BV + lenalidomide, the objective response rate (ORR) was 56.7% (73.3% in CD30+ patients). The median duration of remission was 13.2 months in patients with a complete response (CR) or partial response (PR) and 11.7 months in patients with a CR, PR, or stable disease (SD) &gt;6 months. The median progression-free survival (PFS) and overall survival (OS) were 10.2 months and 14.3 months, respectively, and results were similar in the CD30+ and CD30 &lt;1% groups (manuscript submitted). The clinical activity and manageable safety profiles of BV, lenalidomide, and rituximab as single agents provide a strong rationale for this combination in multiple relapsed and heavily pretreated patients with R/R DLBCL. Study Design and Methods ECHELON-3 (NCT04404283) is a randomized, double-blind, placebo-controlled, active-comparator, multicenter phase 3 study designed to evaluate the efficacy of BV versus placebo, in combination with lenalidomide and rituximab, in patients with R/R DLBCL. Key eligibility criteria will include patients ≥18 years of age with R/R DLBCL with an eligible subtype, ≥2 prior lines of systemic therapy and ineligible for stem cell transplant or CAR-T therapy, Eastern Cooperative Oncology Group performance status 0 to 2, fluorodeoxyglucose-avid disease by positron emission tomography (PET), and bidimensional measurable disease of at least 1.5 cm by computed tomography (CT). Patients with a history of another malignancy within the past 2 years, any uncontrolled Grade ≥3 infection, Grade ≥2 peripheral neuropathy, or previous exposure to BV or lenalidomide will not be eligible. After completing the safety run-in period, patients (n=400) will be randomized 1:1 to receive either BV (1.2 mg/kg intravenously every 3 weeks [Q3W]) or placebo (intravenously Q3W) in combination with lenalidomide (20 mg orally daily) and rituximab (1400 mg subcutaneously Q3W from Cycle 2, with 375 mg/m 2 intravenously on Cycle 1 Day 1). Treatment may continue while there is clinical benefit (SD or better) without progression or unacceptable toxicity. Patients will be stratified by CD30 expression (≥1% [positive] vs &lt;1% [negative]), prior allogeneic or autologous stem cell transplant therapy (received or not), prior CAR-T therapy (received or not), and cell of origin (germinal center B-cell [GCB] or non-GCB). The dual primary endpoints are PFS per blinded independent central review (BICR) in the intention-to-treat (ITT) and CD30+ populations. Key secondary endpoints are OS in the ITT and CD30+ populations, and ORR per BICR. Other secondary endpoints include CR rate, duration of objective response, and safety and tolerability of the combination. Disease response will be assessed by BICR and the investigator according to the Lugano Classification Revised Staging System for nodal non-Hodgkin and Hodgkin lymphomas (Cheson 2014). Radiographic disease evaluations, including contrast-enhanced CT and PET, will be assessed at baseline, then every 6 weeks from randomization until Week 48, then every 12 weeks thereafter. PET is not required after CR is achieved For the analyses of PFS per BICR, the stratified log-rank test will be used to compare PFS between the 2 treatment groups. Hazard ratios will be estimated using the stratified Cox regression model. PFS will also be summarized using the Kaplan-Meier method. Similar methods will be used to estimate OS and other time-to-event efficacy endpoints. The study is currently enrolling and will be open in 16 countries. Disclosures Bartlett: Merck: Research Funding; Kite, a Gilead Company: Research Funding; Janssen: Research Funding; Genentech: Research Funding; Forty Seven: Research Funding; Celgene: Research Funding; Bristol Myers Squibb: Research Funding; Autolus: Research Funding; Seagen: Consultancy, Research Funding; Roche/Genentech: Consultancy; ADC Therapeutics: Consultancy, Research Funding; Millennium: Research Funding; Pharmacyclics: Research Funding. Yasenchak: Seagen Inc.: Research Funding. Harwin: Sarah Cannon Research Institute: Other: Grants, Research Funding. Sims: Seagen Inc.: Current Employment. Nowakowski: Celgene, MorphoSys, Genentech, Selvita, Debiopharm Group, Kite/Gilead: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene, NanoString Technologies, MorphoSys: Research Funding.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1950-1950 ◽  
Author(s):  
John A Lust ◽  
Charles Barranco ◽  
Saad Z Usmani ◽  
Frits van Rhee ◽  
Mehdi Hamadani ◽  
...  

Abstract Eukaryotic translation initiation factor 5A (eIF5A) has been implicated in the regulation of cell proliferation, apoptosis, and inflammation, and is the only known protein to be modified by hypusination. Hypusinated eIF5A, the predominant form of eIF5A in cancer cells, is involved in cell survival and activation of inflammatory pathways. In contrast, accumulation of the unhypusinated form of eIF5A is associated with apoptosis and mutants of eIF5A that cannot be hypusinated (e.g. eIF5AK50R) are pro-apoptotic. SNS01-T was designed to treat B-cell cancers and consists of two active components: a plasmid DNA expressing the pro-apoptotic eIF5AK50R under the control of a B cell-specific promoter, and an siRNA against an untranslated region of native eIF5A mRNA. When these two components are combined with linear polyethyleneimine (PEI), the nucleic acids are condensed into nanoparticles for protection from degradation in the blood and enhanced cellular delivery. The mode of action of SNS01-T is siRNA-mediated inhibition of hypusinated eIF5A and simultaneous over-expression of pro-apoptotic eIF5AK50R to induce cell death. In vitro cell studies and in vivo xenograft studies have demonstrated the efficacy of this approach. The safety and tolerability of intravenous administration of SNS01-T is being investigated in a first-in-human Phase1b/2a study in patients with relapsed or refractory multiple myeloma (MM), mantle cell lymphoma (MCL) or diffuse large B cell lymphoma (DLBCL). Eligible patients are being enrolled sequentially into four cohorts at increasing doses. Each patient receives an intravenous infusion of SNS01-T twice weekly for 6 consecutive weeks. Eligible patients must have been diagnosed with MM according to IMWG criteria, or with MCL or DLBCL with histologic confirmation. Patients also must have measurable disease, have relapsed or refractory disease after two or more prior treatment regimens, have a life expectancy of at least 3 months, and not be eligible to receive any other standard therapy known to extend life expectancy. The primary objective is to evaluate the safety and tolerability of multiple escalating doses of SNS01-T. Secondary objectives include analysis of pharmacokinetics, immunogenicity, pro-inflammatory cytokines, and therapeutic efficacy. The required 3 patients per cohort have completed the dosing schedule in cohorts 1 and 2 from a total of 10 patients enrolled (9 patients with MM and 1 with DLBCL). Of the ten patients enrolled, four completed the full treatment period, two did not complete dosing but were evaluable for safety, and four (three in cohort 1 and one in cohort 2) discontinued treatment after fewer than 8 doses and were not evaluable. There were no drug-related serious adverse events or dose limiting toxicities in either cohort 1 or 2. In cohort 1 (0.0125 mg/kg SNS01-T), two of three evaluable patients did not progress on treatment and were considered stable at week 3 and week 6, the end of the dosing regimen. The third patient progressed after receiving 10 of the 12 doses and was evaluable for safety. In cohort 2 (0.05 mg/kg), 3 patients (2 with MM and 1 with DLBCL) were evaluable for safety. Stabilization of serum monoclonal protein levels was observed in one MM patient of cohort 2. Two patients (1 with MM and 1 with DLBCL) progressed after receiving 8 of the 12 doses and were evaluable for safety. Results from ongoing pharmacokinetic studies, immunogenicity studies, and quantification of pro-inflammatory cytokines will be discussed. The planned dose levels for the third and fourth groups are 0.2 and 0.375 mg/kg, respectively. The results to date of this first-in-human clinical trial indicate that SNS01-T can be administered safely and the MTD has not yet been reached (Clinical Trials.gov Identifier: NCT01435720). Disclosures: Barranco: Senesco Technologies: Consultancy. Usmani:Celgene, Onyx, Millenium: Consultancy, Research Funding, Speakers Bureau. van Rhee:Jansen&Jansen: Research Funding. Thompson:Senesco Technologies: Consultancy, Equity Ownership, Membership on an entity’s Board of Directors or advisory committees, Research Funding. Taylor:Senesco Technologies: stock options Other. Dondero:Senesco Technologies: Employment. Browne:Senesco Technologies Inc.: Employment, Equity Ownership, Membership on an entity’s Board of Directors or advisory committees. Siegel:Celgene, Millenium, Onyx (same for all): Honoraria, Membership on an entity’s Board of Directors or advisory committees, Speakers Bureau.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 998-998 ◽  
Author(s):  
Sattva S. Neelapu ◽  
Frederick L. Locke ◽  
Nancy L Bartlett ◽  
Tanya Siddiqi ◽  
Caron A. Jacobson ◽  
...  

Abstract Background: ZUMA-1 is a phase 1-2 multicenter, open-label study evaluating the safety and efficacy of KTE-C19 in patients with refractory aggressive B-cell non-Hodgkin lymphoma. In phase 1, KTE-C19 demonstrated ongoing complete remissions with a tolerable safety profile (Neelapu, ASCO 2016). The phase 2 portion of the study has 2 cohorts based on tumor type: diffuse large B-cell lymphoma (cohort 1) or PMBCL/TFL (cohort 2). Results from cohort 2 at the time of the first pre-specified interim analysis of ZUMA-1 are presented here. Methods: Patients received KTE-C19 at a target dose of 2 × 106 (minimum 1 × 106) anti-CD19 chimeric antigen receptor (CAR) T cells/kg after a low-dose conditioning chemotherapy regimen of cyclophosphamide (500 mg/m2) and fludarabine (30 mg/m2) daily for 3 days. The primary endpoint was overall remission rate per International Working Group criteria (Cheson, J Clin Oncol2007). Key secondary endpoints include duration of response, incidence of adverse events (AEs), levels of CAR T cells in the blood, and levels of serum cytokines. Key inclusion criteria include ≥ 18 years old, Eastern Cooperative Oncology Group (ECOG) performance status 0-1, and chemorefractory disease defined as progressive disease or stable disease as best response to last line of therapy, or disease progression ≤12 months after autologous stem cell transplant. Patients must have received prior anti-CD20 therapy and an anthracycline-containing regimen. Results: As of June 16, 2016, six patients in cohort 2 were treated with KTE-C19 and had at least 30 days of follow up. The median follow up time was 3.2 months. Fifty percent of patients had PMBCL and 50% had TFL. Median age was 55 years (range, 28-60), 67% were male, 67% ECOG performance status 0, and 50% were refractory to second or greater line of chemotherapy while 50% relapsed after autologous stem cell transplant. The objective response rate was 100%. All patients have ongoing complete remissions. Worst grades 3 and 4 treatment-emergent AEs occurred in 17% and 67% of patients, respectively. There were no grade 5 events. All grade 4 treatment-emergent AEs were cytopenias. KTE-C19-related worst grade 3 and 4 AEs occurred in 50% and 17% of patients, respectively. Patient incidence of any grade/grade 3/grade 4 CRS and neurotoxicity were 100%/0%/0% and 67%/33%/0%, respectively. All KTE-C19-related AEs have resolved. Biomarker endpoints will be presented. Conclusions: KTE-C19 demonstrated early promising activity in patients with refractory PMBCL and TFL. The predominant toxicities of CRS and neurotoxicity were generally reversible. Updated trial results from 11 patients at interim analysis 2 will be presented. Clinical trial: NCT02348216. This study is supported in part by funding from The Leukemia & Lymphoma Society (LLS) Therapy Acceleration Program® Disclosures Locke: Kite: Membership on an entity's Board of Directors or advisory committees. Bartlett:Gilead: Consultancy. Siddiqi:Pharmacyclics, LLC, an AbbVie Company: Speakers Bureau; Janssen: Speakers Bureau; Seattle Genetics: Speakers Bureau; Kite pharma: Other: Funded travel, 1 day registration, and 1 night hotel stay for EHA2016 so I could present trial data there. . Jacobson:Kite: Membership on an entity's Board of Directors or advisory committees. Westin:ProNAi: Membership on an entity's Board of Directors or advisory committees; Spectrum: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Chugai: Membership on an entity's Board of Directors or advisory committees. Chavez:Janssen: Speakers Bureau. LaCasce:Seattle Genetics: Consultancy; Seattle Genetics: Consultancy; Forty Seven: Consultancy; Forty Seven: Consultancy. Bot:Kite Pharma: Employment, Equity Ownership. Rossi:Kite Pharma: Employment, Equity Ownership. Jiang:Kite Pharma: Employment, Equity Ownership. Aycock:Kite Pharma: Employment, Equity Ownership. Elias:Kite: Employment, Equity Ownership. Wiezorek:Kite Pharma: Employment, Equity Ownership. Go:Kite Pharma: Employment, Equity Ownership.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5115-5115
Author(s):  
Amy Sharma ◽  
Sadia Riaz ◽  
Jonathan E. Kolitz ◽  
Jacqueline C. Barrientos ◽  
Steven L Allen

Abstract Introduction Large cell lymphoma transformed from an indolent lymphoproliferative disorder typically carries a worse prognosis than de novo diffuse large B cell lymphoma. When transformation to large cell lymphoma occurs in CLL (Richter's syndrome), traditional anthracycline or platinum based therapy is associated with a median survival of <12 months. Better, more targeted therapies are needed. We describe 4 patients with transformation to large cell lymphoma who responded to ibrutinib. Cases: Patient A, age 68 at transformation, was a 64 year old male at diagnosis with CLL Rai stage 1. He was initially asymptomatic with a performance status of 0. 4 years later he developed dyspnea on exertion after one block and was found to have a left pleural effusion with diffuse lymphadenopathy with increased PET avidity. Biopsy of a supraclavicular node was positive for extracavitary primary effusion lymphoma, HHV8+, CD5-, CD10-. Patient was given R-CHOP x 6 cycles; he relapsed after 18 months and was given ibrutinib 560mg daily with monthly rituximab x 6 and achieved a PR with reversion to CLL. He is currently continuing ibrutinib in this remission for 10+ months. Patient B, age 90 at transformation, was a 68 year old female at diagnosis of CLL, Rai stage 0. She developed stage III CLL 18 years after diagnosis, was treated with BR x 6 cycles. 2 years later she developed Richter's transformation which was CD10+. Although she achieved a PR after 4 months of ibrutinib 560mg with monthly rituximab, her PS was 4 and she was transferred to hospice and expired 4.5 months after initiating ibrutinib/rituximab. Patient C, age 87 at relapse, was a 73 year old male at diagnosis when he originally presented with stage 1 DLBCL transformed from marginal zone lymphoma. He had 3 cycles of R-CHOP and RT to involved area and was disease free for 14 years until he had worsening thrombocytopenia. This was monitored for 3 years until age 87 when CT/PET showed increasing SUV in multiple lymph nodes and the spleen. Biopsy showed diffuse large B cell lymphoma, CD10-. He was started on ibrutinib 560mg with monthly rituximab x 6. He achieved a CR by CT/PET except for persistent splenic disease. He underwent splenectomy and continues in CR on ibrutinib at 9+ months. Patient D is an 83 year old female with large cell transformation from marginal zone lymphoma at diagnosis. She had stage IV disease with large cells involving pleural fluid and bone marrow. She was CD10-. She received R-CHOP x 3 with progressive disease. At that time ibrutinib 560mg alone was initiated. She has a CR based on recent CT/PET findings and is continuing ibrutinib at 18+ months. Conclusion: All of the above patients responded to ibrutinib given with or without rituximab with symptomatic and objective remissions; all of the CD10 negative cases are alive and still responding 9-18 months after initiating therapy. Studies examining the efficacy of ibrutinib in diffuse large B cell lymphoma are underway. This report supports the need for further study of ibrutinib in the transformed setting, particularly in the elderly where patients may not be appropriate for aggressive therapies. Disclosures Off Label Use: Ibrutinib was used to treat transformed large cell lymphoma.. Kolitz:Pharmacyclics: Membership on an entity's Board of Directors or advisory committees. Barrientos:Gilead: Research Funding; NIH/NCATS: Research Funding; ASH-AMFDP: Research Funding. Allen:Millennium: Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Equity Ownership; Onconova: Membership on an entity's Board of Directors or advisory committees; Alexion: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2502-2502
Author(s):  
Alex Iukuridze ◽  
Jennifer Berano Teh ◽  
Justin Ramos ◽  
Teresa Cabrera Vera ◽  
Kyuwan Lee ◽  
...  

Abstract Introduction: Chimeric antigen receptor T cell (CAR-T) therapy is an effective treatment for patients with relapsed/refractory diffuse large B cell lymphoma (DLBCL) or other B-cell lymphomas. However, the potent anti-lymphoma effect of CAR-T is balanced by the risk of acute toxicities, namely cytokine release syndrome (CRS) and Immune effector cell-Associated Neurotoxicity Syndrome (ICANS), as well as the variable length of progression-free survival (PFS) after CAR-T. Tools to better risk-stratify for adverse outcomes and to guide targeted interventions are lacking. Sarcopenia (loss of lean muscle mass) is an important cause of age-related functional decline in the general population and is an independent predictor of health outcomes in patients with solid and hematologic cancers, irrespective of age or comorbidity. Advances in software technology have facilitated the near real-time integration of body composition measurements into imaging studies obtained as part of standard clinical care. To date, there have been no studies to examine the association between sarcopenia and outcomes after CAR-T therapy. Methods: Using a retrospective cohort design, 280 consecutive patients with DLBCL or B-cell lymphoma, age ≥18y, and treated with CAR-T therapy between 2015 to 2020 at a single center were included in the study. This analysis was restricted to 226 (80.7%) patients with available computed tomography scans ≤60d from CAR-T. Skeletal muscle area was ascertained from abdominal scans using an automatic image analysis software (APACS; Voronoi Health Analytics; Vancouver, Canada); 3rd lumbar vertebra was used as a landmark because of its high correlation with whole-body muscle mass (J Clin Oncol 2016 34:1339); Figure. Trained researchers blinded to patient demographics and outcomes manually validated these measurements (SliceOmatic; Tomovision; Quebec, Canada). Skeletal muscle index (SMI) was calculated as the ratio of skeletal muscle area (cm 2) divided by height (m). Sarcopenia was defined according to sex-based cutoffs (lowest SMI tertile). Kaplan-Meier method was used to examine PFS at one-year. Multivariable regression was used to calculate the hazard ratio (HR) for PFS and odds ratio (OR) for toxicities with 95% confidence intervals (CI), adjusted for covariates (demographics [age, race/ethnicity], disease characteristics [largest lymph node diameter, blood lactate dehydrogenase], CAR-T product, ECOG performance status). Results: Median age at CAR-T was 63y (range: 18-84); 65.9% were male; 50.9% were non-Hispanic white; 8.8% had ECOG ≥2; 80.5% had a diagnosis of DLBCL; CAR-T products: axicabtagene ciloleucel (51.3%), lisocabtagene maraleucel (31.9%), other (16.8%); 46.9% were treated on a clinical trial; median residual lymph node diameter prior to CAR-T was 2.3cm (range: 0-17.2); 8.0% underwent HCT &lt;1 year after CAR-T and follow-up was censored at HCT. Outcomes: 59.1% developed CRS (18.2% grade ≥2) and 30.1% developed ICANS (15.9% grade ≥2). In adjusted analyses, the odds of developing CRS or ICANS was 1.9-fold (CRS: 1.89 [95%CI: 1.02-3.5], ICANS: 1.93 [1.06-3.51]) higher among patients who were sarcopenic (reference: normal body composition). Average length of hospitalization was also longer (25.6d vs. 21.9d; p=0.037) among patients with sarcopenia. Survival: One-year PFS for the overall cohort was 50.1% (±4.2); PFS was significantly worse for patients who were sarcopenic compared to those with normal muscle mass (35.1% [±6.2] vs. 57.7% [±4.3], p=0.005; Figure). In adjusted analyses, sarcopenia was associated with inferior one-year PFS (HR=1.73 [CI: 1.12-2.68]) compared to those with normal muscle mass. Conclusion: Sarcopenia is an important and independent predictor of outcomes after CAR-T with potential downstream health-economic consequences, including increased burden of acute toxicities and prolonged length of hospitalization. Taken together, these data form the basis for real-time decision making prior to CAR-T (e.g. pre-habilitation, consideration of alternative treatments), or during/shortly after CAR-T (e.g. increased supportive care, rehabilitation), setting the stage for innovative strategies to improve outcomes after CAR-T therapy. Figure 1 Figure 1. Disclosures Artz: Radiology Partners: Other: Spouse has equity interest in Radiology Partners, a private radiology physician practice. Budde: Merck, Inc: Research Funding; Amgen: Research Funding; Astra Zeneca: Research Funding; Mustang Bio: Research Funding; Novartis: Consultancy; Gilead: Consultancy; Roche: Consultancy; Beigene: Consultancy. Herrera: Merck: Consultancy, Research Funding; Gilead Sciences: Research Funding; Bristol Myers Squibb: Consultancy, Research Funding; AstraZeneca: Consultancy, Research Funding; Karyopharm: Consultancy; Kite, a Gilead Company: Research Funding; Seagen: Consultancy, Research Funding; ADC Therapeutics: Consultancy, Research Funding; Takeda: Consultancy; Tubulis: Consultancy; Genentech: Consultancy, Research Funding. Popplewell: Novartis: Other: Travel; Pfizer: Other: Travel; Hoffman La Roche: Other: Food. Shouse: Kite Pharmaceuticals: Speakers Bureau; Beigene Pharmaceuticals: Honoraria. Siddiqi: Kite Pharma: Membership on an entity's Board of Directors or advisory committees; Juno therapeutics: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; BMS: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Abbvie: Membership on an entity's Board of Directors or advisory committees; AstraZeneca: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; BeiGene: Other: DSM Member, Speakers Bureau; PCYC: Speakers Bureau; Jannsen: Speakers Bureau; Dava Oncology: Honoraria; ResearchToPractice: Honoraria. Forman: Lixte Biotechnology: Consultancy, Current holder of individual stocks in a privately-held company; Allogene: Consultancy; Mustang Bio: Consultancy, Current holder of individual stocks in a privately-held company.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3851-3851
Author(s):  
Audrey M. Sigmund ◽  
Nathan Denlinger ◽  
Amneet Bajwa ◽  
Patrick Elder ◽  
David A. Bond ◽  
...  

Abstract Introduction: Outcomes of patients with large B-cell lymphoma that relapse after frontline anthracycline based chemotherapy are typically poor, with a 3-year event-free survival of approximately 30% (Gisselbrecht JCO 2010). Chimeric antigen receptor T-cell (CAR-T) therapy represents a breakthrough therapy for these patients, with an overall response rate (ORR) of 83% and a complete response (CR) rate of 58% for axicabtagene ciloleucel (axi-cel) seen in the ZUMA-1 trial, with similar rates for tisagenlecleucel in the JULIET trial (Locke Lancet Oncol 2018; Schuster NEJM 2018). Unfortunately, the majority of patients treated with CAR-T therapy experience disease progression. There is limited data evaluating the best salvage regimen for these patients. Thus, we sought to assess outcomes in large B-cell lymphoma patients with progressive disease post CAR-T cell therapy with the goal of identifying those therapies with optimal outcomes. Methods: A retrospective study was performed on all patients with large B-cell lymphoma undergoing leukapheresis for CAR-T therapy (tisagenlecleucel or axi-cel) at the Ohio State University from December 2017 to January 2021. Patients who died prior to CAR-T infusion were excluded from analysis. Demographics and disease characteristics as well as best response to CAR-T and date of relapse or progression following therapy were collected. First salvage therapy at relapse or progression and response to therapy were also collected, with choice of therapy driven by the treating physician. Patients were divided by salvage regimen into five groups for analysis: checkpoint inhibitor based, lenalidomide based, Bruton Tyrosine Kinase inhibitor (BTKi), chemoimmunotherapy, and other (including small molecular inhibitor, radiation, allogeneic stem cell transplant, antibody drug conjugates, and bispecific antibodies). The primary endpoint was overall survival (OS), which was calculated using Kaplan Meier Curves. Rates of CR and ORR were also assessed. Results: A total of 144 patients underwent leukapheresis for CAR-T cell therapy during the time period; of these patients, 13 died prior to undergoing CAR-T cell infusion and were excluded from analysis. The primary cohort included 131 patients. Median age at the time of T-cell collection was 62 years old (range 23-85) and 61% were male. The majority (50%) had germinal center (GCB) subtype, with 42% non-GCB and subtype unavailable for 8%. A small number (3%) had primary mediastinal B-cell lymphoma. The majority had high-risk disease, with 45% having primary refractory disease, 14.5% double or triple hit, and 81% Ann Arbor stage III or IV at diagnosis. Median prior lines of therapy was 3 (range 0-10). Sixty-six patients received axi-cel and 65 patients received tisagenlecleucel. Forty percent of patients attained a CR, 18% partial response (PR), 3% stable disease (SD), 33% progressive disease (PD), and 6% of patients died prior to disease assessment. For those 76 patients that received a CR or PR to therapy, 43% relapsed post CAR-T and 57% remained in CR at last follow-up. Of those patients who relapsed or progressed post CAR-T, 69% (54/78) patients received additional therapy. The most common therapies utilized were lenalidomide based (35%), BTKi (22%), chemoimmunotherapy (13%), and checkpoint inhibitor based (15%). Other therapies represented 15% of cases and included small molecular inhibitor, radiation, allogeneic stem cell transplant, antibody drug conjugates, and bispecific antibodies (Table 1). Overall response rates and median OS for the groups were 50% and 2.25 years for BTKi, 13% and 0.96 years for checkpoint inhibitor based, 71% and not reached (NR) for chemoimmunotherapy, 47% and 2.4 years for lenalidomide based, and 75% and NR for other (Table 1; Figure 1). Median OS for those patients who did not receive any salvage therapy was 0.19 years. Conclusion: Consistent with prior studies, median OS following relapse post CAR-T therapy was poor, with median OS 0.19 years for patients who did not receive therapy and ranging from 0.96 years to NR for those that did. Rates of CR were highest in patients treated with BTKis. In our series, ORR rate to checkpoint inhibitors was relatively low in contrast to other recently published retrospective reports. Future prospective studies are needed to further assess the optimal therapy for patients who relapse post CAR-T therapy. Figure 1 Figure 1. Disclosures Bond: Kite/Gilead: Honoraria. Brammer: Seattle Genetics: Speakers Bureau; Kymera Therapeutics: Consultancy; Celgene: Research Funding. de Lima: Miltenyi Biotec: Research Funding; Incyte: Membership on an entity's Board of Directors or advisory committees; BMS: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees. Jaglowski: Novartis: Consultancy, Research Funding; Takeda: Consultancy; Juno: Consultancy; Kite, a Gilead Company: Consultancy, Research Funding; CRISPR Therapeutics: Consultancy. Kittai: Bristol-Meyers Squibb: Consultancy; Janssen: Consultancy; Abbvie: Consultancy.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3238-3238 ◽  
Author(s):  
Brandon Imber ◽  
M. Lia Palomba ◽  
Carl DeSelm ◽  
Connie Lee Batlevi ◽  
Parastoo B. Dahi ◽  
...  

Background: CD19-targeted chimeric antigen receptor T cell (CAR T) therapies have remarkable overall response rates (ORR) for relapsed diffuse large B cell lymphoma (DLBCL). There is strong rationale to use a radiotherapy (RT) bridge during the cell manufacturing process including palliation, local control and cytoreduction with limited count impact. Recent data from our institution suggests RT may augment an immune response and sensitize antigen negative cells to CAR-mediated death. This series details our early experience using RT conditioning. Methods: 13 patients (median age 64 years) with DLBCL (n=9) or transformed follicular lymphoma (n=4) were analyzed. Overall, patients had a median of 2 prior therapies (range 1-8) including 3 with autologous transplant, 3 with distant RT and 1 with CAR T infusion. Several CAR products were used, including axicabtagene ciloleucel (n=8), JCAR017 (n=3, per NCT02631044), tisagenlecleucel (n=1) and EGFRt/19-28z/4-1BBL "armored" CAR (n=1, per NCT03085173). Most patients (n=10) began RT post apheresis with median duration between RT and CAR infusion of 20d (range 13-80, Figure 2). The most common RT regimen (n=8) was 20 Gy in 5 fractions (range 20-47 Gy) but 2 received our pre-transplant regimen of 30 Gy in 20 BID fractions. None received concurrent chemotherapy with RT but one had a cycle post RT and pre CAR. All had cyclophosphamide and fludarabine lymphodepletion. PET response was evaluated by Lugano criteria. Results: Three patients had limited stage PET avid disease at RT and were treated comprehensively pre-CAR. The remaining 10 were advanced stage and were treated palliatively to limited sites. Irradiated sites included the pelvis/groin (n=4), neck (n=3), intraabdominal (n=2) and extremity (n=2). Most (n=10) had intensity modulated radiotherapy. RT fields were large (median planning treatment volume of 887 cc, range 163-1641). Post RT PET interpretation was challenging given a short interval since RT ended (median 11d) but of 11 evaluable patients, many (n=8, 73%) had partial response (PR). Though locally controlled, most (n=10, 91%) had out of field progressive disease (PD) pre-CAR. Post CAR T, no severe adverse events in the RT field were noted, 9/13 had cytokine release syndrome (n=1 grade 3, n=2 grade 2) and 4 had neurotoxicity (n=3 grade 3). At day 30, ORR was 90%; of 10 evaluable patients, 7 had complete response (CR) and 2 had partial response (PR). Of the 7 evaluable patients at day 90, 4 (57%) had continued CR and the other 3 (43%) had PD and subsequently died from DLBCL. One relapsed at 95d post armored CAR both in and out of the RT field, and the other relapsed at 64d post JCAR017 primarily out of field. Conclusions: Use of RT as a CAR T bridging strategy is feasible and associated with excellent pre-CAR local control and initial post CAR ORR in a cohort of heavily pre-treated DLBCL patients. We observed moderate serious CAR toxicity that did not appear to be augmented by RT. Future efforts should clarify the optimal RT timing/dose and assess the potential for incremental immunogenicity with combined therapy. Disclosures Palomba: Hemedicus: Other: Immediate Family Member, Speakers Bureau ; Merck & Co Inc.: Other: Immediate Family Member, Consultancy (includes expert testimony); Seres Therapeutics: Other: Immediate Family Member, Equity Ownership and Membership on an entity's Board of Directors or advisory committees; STRAXIMM: Other: Immediate Family Member, Membership on an entity's Board of Directors or advisory committees; Kite Pharmaceuticals: Other: Immediate Family Member, Membership on an entity's Board of Directors or advisory committees; Pharmacyclics: Membership on an entity's Board of Directors or advisory committees; Noble Insights: Consultancy; Evelo: Other: Immediate family member, Equity Ownership; MSK (IP for Juno and Seres): Other: Immediate Family Member, Patents & Royalties - describe: intellectual property rights . Batlevi:Juno Therapeutics: Consultancy, Membership on an entity's Board of Directors or advisory committees. Giralt:Celgene: Consultancy, Research Funding; Takeda: Consultancy; Sanofi: Consultancy, Research Funding; Amgen: Consultancy, Research Funding. Noy:Medscape: Honoraria; Prime Oncology: Honoraria; Pharamcyclics: Research Funding; Raphael Pharma: Research Funding; NIH: Research Funding; Janssen: Consultancy. Park:Amgen: Consultancy; Autolus: Consultancy; AstraZeneca: Consultancy; GSK: Consultancy; Incyte: Consultancy; Kite Pharma: Consultancy; Novartis: Consultancy; Takeda: Consultancy; Allogene: Consultancy. Sauter:Juno Therapeutics: Consultancy, Research Funding; Genmab: Consultancy; GSK: Consultancy; Spectrum Pharmaceuticals: Consultancy; Novartis: Consultancy; Celgene: Consultancy; Kite/Gilead: Consultancy; Precision Biosciences: Consultancy; Sanofi-Genzyme: Consultancy, Research Funding. Scordo:Angiocrine Bioscience, Inc.: Consultancy; McKinsey & Company: Consultancy. Shah:Janssen: Research Funding; Amgen: Research Funding. Sadelain:Memorial Sloan Kettering Cancer Center: Employment; Juno Therapeutics: Consultancy, Patents & Royalties, Research Funding; Fate Therapeutics: Consultancy, Patents & Royalties. Perales:Bellicum: Honoraria, Membership on an entity's Board of Directors or advisory committees; Bristol-Meyers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; Incyte: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Nektar Therapeutics: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Omeros: Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; Merck: Consultancy, Honoraria; MolMed: Membership on an entity's Board of Directors or advisory committees; NexImmune: Membership on an entity's Board of Directors or advisory committees; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees; Medigene: Membership on an entity's Board of Directors or advisory committees; Servier: Membership on an entity's Board of Directors or advisory committees; Kyte/Gilead: Research Funding; Miltenyi: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document