Safety and Efficacy Of Obinutuzumab (GA101) Plus CHOP Chemotherapy In First-Line Advanced Diffuse Large B-Cell Lymphoma: Results From The Phase 2 Gather Study (GAO4915g)

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1820-1820 ◽  
Author(s):  
Andrew D Zelenetz ◽  
Mehrdad Mobasher ◽  
Luciano J Costa ◽  
Ian Flinn ◽  
Christopher R. Flowers ◽  
...  

Abstract Introduction Diffuse large B-cell lymphoma (DLBCL) is the most common non-Hodgkin lymphoma and has an aggressive natural history. Rituximab (R), a type I anti-CD20 monoclonal antibody (mAb), plus CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone), has improved patient (pt) survival and established a new standard of care. Nonetheless, 35%–40% of pts will relapse during/after first-line treatment and of these, <25% are cured with second-line therapy. Thus, there is a medical need to further improve cure rates, particularly in the first-line setting. Obinutuzumab (GA101; G) is a novel, glycoengineered type II anti-CD20 mAb with increased direct cell death and antibody-dependent cell-mediated cytotoxicity relative to R. Clinical activity of G in R/R DLBCL was confirmed in phase 1/2 studies. Phase 1b studies in indolent lymphoma found G-CHOP to be safe and effective. GATHER is a phase 2, open-label, multicenter study examining the safety and efficacy of G-CHOP in the first-line treatment of advanced DLBCL. Methods Pts with untreated CD20-positive DLBCL, clinical stage (CS) IIX (mass>7.5 cm), III, and IV, and International Prognostic Index (IPI) ≥2 (or any IPI if bulky disease) with measurable disease were treated with standard CHOP (day [d]1, 21-day cycles [c] 1–6) and G (1000 mg intravenously [IV]; d1, 21-d c 1–8 plus additional doses on d8 and d15 of c1). Prophylactic GCSF was not mandatory but allowed per ASCO guidelines. After the safety of regular-infusion G was established in the first 20 pts, Shorter Duration of Infusion (SDI) of G (SDI 120 min and SDI 90 min) was tested. Disease responses were assessed using FDG-PET and CT scans, according to the Cheson 2007 criteria, 6–8 weeks after completion of treatment. Efficacy endpoints were investigator- and independent central review facility-assessed overall response rate (ORR) and complete response (CR). G-CHOP Safety was measured by the incidence and severity of adverse events (AEs) and serious AEs and SDI safety by the incidence of grade (gr) 3/4 infusion-related (IR) AEs (any G-related AE during or within 24 h after infusion). Cell of origin and prognostic marker assessment was performed using mRNA (fluidigm microfluidic dynamic arrays) from baseline tumor samples. Results Eighty pts were enrolled in 13 months: 59% males; median age 60.5 years (range, 24–80); CS IIX 14%, CS III 35%, CS IV 51%; IPI low risk/low-intermediate risk 54%, high-intermediate risk/high risk 46%. Most AEs were gr 1/2. Gr ≥3 and serious AEs with highest frequency are summarized (Fig). IR AEs were in 51 pts (64%), most commonly in c1d1 (48 pts). The majority of IR AEs were gr 1/2; only 2 gr 3 IR AEs occurred, both in c1 d1. There were no gr ≥4 IR AEs. 288 SDI infusions (120-min in 24, 90-min in 264) were administered and associated with 3 gr 1/2 IR AEs and no gr ≥3 IR AEs. G-CHOP was delayed >5 d in 3/79 (4%) in c2, 7/78 (9%) in c3, 8/78 (10%) in c4, 5/77 (6.5%) in c5, and 7/74 (9.5%) in c6. Early treatment discontinuation occurred in 11 pts; 5 for failure to resolve an AE in ≤14 d. The median overall dose intensity of chemo agents across all 6 cycles was 100% (range 17%–104%). The ORR was 66/80 (83%) (44/80 [55%] CR, 22/80 [28%] PR); 1 pt was not evaluable, and 4 had missing data. Nine pts had progressive disease at the end of treatment, and there were 3 deaths from disease progression. Response assessment by an independent central review facility and analysis of efficacy in prognostic molecular subgroups (including cell of origin) will be presented. Conclusions G-CHOP was safely administered to pts with newly diagnosed DLBCL. Dose intensity of CHOP was maintained throughout treatment. Manageable, mild, and moderate IR AEs were frequent in the first cycle of G, but all pts continued the infusion after symptom resolution. Nonetheless, SDI of G was safe for all eligible pts. Neutropenia and febrile neutropenia appear to be the most important AEs. However, these could be addressed by prophylactic administration of G-CSF support. Preliminary efficacy is promising, but it is too early to evaluate progression-free and overall survival. The data provide further rationale for the ongoing randomized phase 3 study of G-CHOP vs R-CHOP in DLBCL. NCT01414855 Disclosures: Zelenetz: Cancer Genetics: Scientific Advisor, Scientific Advisor Other; Genentech, GSK, Roche: Research Funding; GSK, Celgene, Cephalon, Gilead, Seattle Genetics, Sanofi-Avenits USA: Consultancy. Off Label Use: GA101 is a novel, glycoengineered, type II anti-CD20 monoclonal antibody that is designed to enhance direct cell death and antibody-dependent cellular cytotoxicity. It is being investigated in chronic lymphocytic leukemia, Non-Hodgkin’s Lymphoma and other hematologic indications. Mobasher:Roche: Ownership of stock options, Ownership of stock options Other; Genentech: Employment. Costa:Genentech: Research Funding. Flinn:Genentech: Research Funding. Flowers:Celgene, Genentech, Oncology: Consultancy; Abbott, Celgene, Millennium/Takeda, Sanofi-Aventis, Spectrum, Janssen: Research Funding. Sandmann:Roche: Stock options, Stock options Other; Genentech, Inc.: Employment. Trunzer:F. Hoffmann-La Roche: Employment, Stock ownership Other. Vignal:F. Hoffmann-La Roche: Employment, Stock ownership Other.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4233-4233
Author(s):  
Ann Cameron Barr ◽  
Subir Goyal ◽  
Christopher R Flowers ◽  
Jean L. Koff

Abstract Background: Autoimmune (AI) disorders, in particular Sjögren's syndrome (SS), rheumatoid arthritis (RA), and systemic lupus erythematosus (SLE), have been increasingly recognized as risk factors for diffuse large B cell lymphoma (DLBCL). It remains controversial whether increased lymphoma risk arises from the diseases themselves or the immunosuppressive agents used to treat them. It is possible that AI-associated lymphomas comprise a distinct lymphoma subset, exhibiting their own characteristic clinical and biologic behavior. In a recent analysis of >5,000 DLBCL patients in a large national database, our group described decreased lymphoma-related survival in patients with concomitant RA and a trend towards decreased survival in patients with SLE (Koff JL, Clin Lymphoma Myeloma Leuk 2018). Unfortunately, this database did not contain data about cell-of-origin subtype, which is highly associated with survival in DLBCL, or immunosuppressive regimen used to treat AI disease. To determine the prevalence of AI-associated disease among DLBCL patients and better characterize this unique population in terms of demographics, clinical features, and survival outcomes, we examined a large institutional database that included more granular information regarding tumor immunohistochemical (IHC) markers and AI disease treatment. Methods: We used an existing clinic-based registry of DLBCLs to identify patients diagnosed with DLBCL between 1989 and 2018 with concomitant AI disease as defined by the American College of Rheumatology. To be included, AI disease diagnosis was required to precede diagnosis of DLBCL by a minimum of six months. Patient age, sex, race, dates of DLBCL and AI diagnoses, stage, International Prognostic Index (IPI) score, poor performance status (ECOG score ≥2), B symptoms, lymphoma treatment strategies, date of progression/relapse, and date of death were identified via chart review. We also examined history of immunosuppressive therapies for AI disorders, AI serum markers such as ANA and rheumatoid factor, DLBCL cell-of-origin subtype by IHC Hans algorithm, and "double-hit" status by fluorescent in situ hybridization testing for translocations of MYC, BCL2, and BCL6. First-line treatments were categorized as: cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP), rituximab-CHOP (R-CHOP), and "other." We analyzed demographics and baseline clinical characteristics for patients with DLBCL and concurrent SS, RA, SLE, or any other AI disease, plotted overall survival (OS) and progression-free survival (PFS) for these groups, and compared median survival times. Results: Upon initial chart review of 1,045 total patients with DLBCL, 60 patients had a preceding diagnosis of AI disease; of these, 35 carried a diagnosis of either SS, RA, or SLE. Patient characteristics are summarized in the Table. Compared to DLBCL patients with other AI diseases, patients with SS, RA, or SLE were more commonly female (73.3% vs 44.4%, p=0.026), more likely to experience B symptoms (46.7% vs 18.5%, p=0.033) and more likely to have a history of methotrexate treatment (36.7% vs 11.1%, p=0.037). All patients received similar first-line DLBCL treatments regardless of AI disease type. Data on IHC results and FISH status was inconsistently documented prior to 2010. AI factors of interest, including serum markers and immunosuppressive therapies, were also not reliably recorded for any era. Compared to patients with SS and RA, there was a trend towards lower OS in patients with SLE and other AI diseases, although this difference was not statistically significant (Figure). PFS was not significantly different between any groups (range, 41.9%-55.0%). Conclusions: In this retrospective study of over 1,000 patients with DLBCL, concomitant AI disease was uncommon. A more complete evaluation of factors impacting survival in AI-associated DLBCL was limited by small sample size and incomplete documentation of key lymphoma and AI disease features in this dataset. The possibility of lower OS for patients with SLE and other AI diseases should be explored in future prospective studies, which may also better capture molecular features of both DLBCL and AI disease. Disclosures Flowers: Pharmacyclics/ Janssen: Consultancy; Gilead: Consultancy; Gilead: Research Funding; Abbvie: Research Funding; Burroughs Wellcome Fund: Research Funding; Janssen Pharmaceutical: Research Funding; Genentech/Roche: Research Funding; Spectrum: Consultancy; V Foundation: Research Funding; Pharmacyclics: Research Funding; TG Therapeutics: Research Funding; OptumRx: Consultancy; Eastern Cooperative Oncology Group: Research Funding; Karyopharm: Consultancy; Genentech/Roche: Consultancy; National Cancer Institute: Research Funding; Denovo Biopharma: Consultancy; Millennium/Takeda: Research Funding; Bayer: Consultancy; Abbvie: Consultancy, Research Funding; Celgene: Research Funding; BeiGene: Research Funding; Acerta: Research Funding.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1874-1874 ◽  
Author(s):  
Tarec Christoffer El-Galaly ◽  
Chan Yoon Cheah ◽  
Mette Dahl Bendtsen ◽  
Gita Thanarasjasingam ◽  
Roopesh Kansara ◽  
...  

Abstract Background: Secondary CNS involvement (SCNS) is a detrimental complication seen in ~5% of patients with diffuse large B-cell lymphoma (DLBCL) treated with modern immunochemotherapy. Data from older series report short survival following SCNS, typically <6 months. However, data in patients that develop SCNS following primary therapy that contains a rituximab-based-regimen as well as the impact of more intensified treatment for SCNS are limited. Aims: The aims of this study were to i) describe the natural history of SCNS in a large cohort of patients treated with immunochemotherapy, and ii) determine prognostic factors after SCNS. Patients and methods: We performed a retrospective study of patients diagnosed with SCNS during or after frontline immunochemotherapy (R-CHOP or equivalently effective regimens). SCNS was defined as new involvement of the CNS (parenchymal, leptomeningeal, and/or eye) in patients without known CNS involvement at the time of first pathologic diagnosis of DLBCL. Patients were identified from local databases and/or regional/national registries in Denmark, Canada (British Columbia), Australia, Israel, US (University of Iowa/Mayo Clinic SPORE), and England (Guy's and St. Thomas' Hospital, London). Clinico-pathologic and treatment characteristics at the time of SCNS were collected from medical records. Results: In total, 281 patients with SCNS diagnosed between 2001 and 2016 were included. Median age at SCNS was 64 (range 20-93) years and male:female ratio was 1.3. SCNS occurred as part of first relapse in 244 (87%) patients and 112 (40%) had documented concurrent systemic disease at the time of SCNS. The median time from initial DLBCL diagnosis to SCNS was 9 months, which was similar for patients treated with (N=76, 27%) or without upfront CNS prophylaxis (N=205, 73%) (10 vs 9 Mo; P=0.3). The median post-SCNS OS was 4 months (interquartile range 2-13) and the 2yr survival rate was 20% (95% CI 15-25) for the entire cohort. Associations between clinicopathologic features, management strategy, and post-SCNS survival are shown in Table 1, which excludes patients who did not receive any treatment against SCNS, patients treated with steroids alone, and a patient with unavailable treatment information (n=43, 15%). In multivariable analysis, performance status >1, concurrent leptomeningeal and parenchymal involvement, SCNS developing before completion of 1st line treatment, and combined systemic and CNS involvement by DLBCL were associated with inferior outcomes. Upfront CNS prophylaxis did not influence post-SCNS OS. High-dose methotrexate (HDMTX) and/or platinum based treatment regimens (i.e. ICE, DHAP, or GDP [+/- IT treatment and/or radiotherapy], N=163) for SCNS were associated with reduced risk of death (HR 0.45 [0.32-0.62, P<0.01]). The 2yr post-SCNS survival for patients treated with HDMTX and/or platinum-based regimens (N=163) was 29% (95% CI 22-37). For patients with isolated parenchymal SCNS, single modality treatment with radiotherapy resulted in 2-yr OS of 19% (95% CI 8-35). For the subgroup of 49 patients treated with HDMTX- and/or platinum-based regimens for isolated SCNS after 1st line DLBCL treatment and with performance status 0 or 1, the 2yr post-SCNS survival was 46% (95% CI 31-59). Overall, 9% of the patients received HDT with ASCT as part of salvage therapy at the time of SCNS. Amongst 36 SCNS patients without systemic involvement and in CR following intensive treatment (HDMTX and/or platinum-based treatments), 11 patients consolidated with HDT had similar outcomes to 25 patients treated without consolidating HDT (P=0.9, Fig 1) Conclusions: Outcomes for patients with SCNS remain poor in this large international cohort of patients from the immunochemotherapy era. Combined parenchymal and leptomeningeal disease, presence of systemic disease concurrent with SCNS, performance status >1, and SCNS developing during first line treatment were independently associated with inferior OS. However, a significant fraction of patients with isolated SCNS after first line DLBCL treatment and with good performance status may achieve long-term remissions after intensive regimens for SCNS. Disclosures El-Galaly: Roche: Consultancy, Other: travel funding. Cheah:Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Gilead Sciences: Membership on an entity's Board of Directors or advisory committees; Janssen-Cilag: Membership on an entity's Board of Directors or advisory committees, Other: Speaker's Bureau. Kansara:Celgene: Honoraria. Connors:Bristol Myers Squib: Research Funding; NanoString Technologies: Research Funding; F Hoffmann-La Roche: Research Funding; Millennium Takeda: Research Funding; Seattle Genetics: Research Funding. Sehn:roche/genentech: Consultancy, Honoraria; amgen: Consultancy, Honoraria; seattle genetics: Consultancy, Honoraria; abbvie: Consultancy, Honoraria; TG therapeutics: Consultancy, Honoraria; celgene: Consultancy, Honoraria; lundbeck: Consultancy, Honoraria; janssen: Consultancy, Honoraria. Opat:Roche: Consultancy, Honoraria, Other: Provision of subsidised drugs, Research Funding. Seymour:Genentech: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie Inc.: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Villa:Celgene: Honoraria; Lundbeck: Honoraria; Roche: Honoraria, Research Funding.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1679-1679
Author(s):  
Paul G. Richardson ◽  
Ruben Niesvizky ◽  
Jay Yang ◽  
Neelu Yadav ◽  
Helen Hsu ◽  
...  

Abstract Background: Multiple myeloma (MM) is a heterogeneous disease of monoclonal plasma cells. More than 40% of patients with MM harbor translocations of the immunoglobulin heavy chain (IGH) gene on chromosome 14, leading to overexpression of putative oncogenes which can ultimately lead to plasma cell-derived, post-malignancy MM. One such translocation, t(4;14), is seen in 15% of patients and juxtaposes IGH control elements with two genes on chromosome 4, FGFR3 and MMSET. Patients harboring the (4;14) translocation have a poor response to standard of care therapies and an overall poor prognosis. MMSET expression has been confirmed as a driver in t(4;14) MM pathogenesis, but MMSET inhibitors have not yet been successfully developed in the clinic. Since MMSET generates the substrate for Su(var)3-9, enhancer of zeste, trithorax domain-containing 2 (SETD2) activity, SETD2 inhibition offers promise for targeting the underlying oncogenic mechanism in t(4;14) MM. Diffuse large B-cell lymphoma (DLBCL) is the most common type of lymphoma in the United States, with a 5-year relative survival rate of 53% in patients diagnosed with stage IV DLBCL. Given the poor survival outcomes and low remission rates for patients with relapsed or refractory (R/R) DLBCL, there is a need for better treatment options. Although SETD2 mutations are described in DLBCL, the mechanism of action of SETD2 inhibitors remains unclear. EZM0414 is a potent and selective, orally bioavailable small-molecule inhibitor of SETD2. Preclinical data have demonstrated antitumor activity of EZM0414 in both t(4;14) and non-t(4;14) MM and DLBCL models. This study (enrollment scheduled to begin Q3 2021) will evaluate the safety and efficacy of EZM0414 when administered as monotherapy in patients with R/R MM with or without t(4;14), or with R/R DLBCL. Study Design and Methods: This first-in-human, 2-part, multicenter, open-label study will enroll patients aged ≥18 years with R/R MM who have received prior treatment with immune modulators, proteasome inhibitors, and anti-CD38 therapy, or who are intolerant to established therapies known to provide clinical benefit in MM, or with R/R DLBCL who have received at least 2 prior lines of therapy, including treatment with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP); rituximab, etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin (R-EPOCH); rituximab, cyclophosphamide, vincristine, doxorubicin, and dexamethasone (R-hyper CVAD); or other standard of care therapies. Patients eligible for autologous stem cell transplantation will be excluded from this study. The first part of the study will be a phase 1 dose escalation study using a Bayesian optimal interval design to evaluate the safety and tolerability of EZM0414 in patients with R/R MM with or without t(4;14), or with R/R DLBCL. Six dose levels of 100, 200, 300, 400, 600, and 900 mg administered once daily will be tested. Patients will be enrolled and treated in a cohort size of 3, and up to 36 patients will be enrolled to evaluate at least 10 patients at the maximum tolerated dose (MTD). The MTD will be selected at the dose level with a target dose-limiting toxicity rate ≤25%. The second part of the study (phase 1b) will be a dose expansion at the MTD in patients with R/R MM with or without t(4;14), or with R/R DLBCL using the Bayesian optimal phase 2 design. Dose expansion will include 3 cohorts of up to 20 patients each. Cohort 1 will enroll patients with t(4;14)-positive R/R MM, cohort 2 will enroll patients with t(4;14)-negative R/R MM, and cohort 3 will enroll patients with R/R DLBCL. The primary endpoints will be determining safety, dose-limiting toxicities, the MTD, and a recommended phase 2 dose. Secondary endpoints include the objective response rate, progression-free survival, and duration of response. Exploratory endpoints include a pharmacokinetic/pharmacodynamic profile analysis and the determination of mechanism of action biomarkers, such as histones and histone methylation. The study design will include a futility assessment in the phase 1b part of the study, which will be initiated when clinical data from the first 10, 15, and 20 enrolled patients in the expansion cohort are available. Disclosures Richardson: Takeda: Consultancy, Research Funding; Karyopharm: Consultancy, Research Funding; Secura Bio: Consultancy; Janssen: Consultancy; GlaxoSmithKline: Consultancy; AstraZeneca: Consultancy; Jazz Pharmaceuticals: Consultancy, Research Funding; Regeneron: Consultancy; Celgene/BMS: Consultancy, Research Funding; AbbVie: Consultancy; Sanofi: Consultancy; Protocol Intelligence: Consultancy; Oncopeptides: Consultancy, Research Funding. Niesvizky: BMS: Consultancy, Research Funding; GSK: Consultancy, Research Funding; Janssen: Consultancy, Research Funding; Karyopharm: Consultancy, Research Funding; Takeda: Consultancy, Research Funding. Yang: Epizyme, Inc.: Current Employment, Other: May own stock/options. Yadav: Epizyme, Inc.: Current Employment, Current holder of stock options in a privately-held company. Hsu: Epizyme, Inc.: Current Employment, Current holder of stock options in a privately-held company. Flowers: Iovance: Research Funding; Adaptimmune: Research Funding; Guardant: Research Funding; Denovo: Consultancy; Celgene: Consultancy, Research Funding; Karyopharm: Consultancy; Kite: Research Funding; Spectrum: Consultancy; Biopharma: Consultancy; SeaGen: Consultancy; Pharmacyclics/Janssen: Consultancy; Epizyme, Inc.: Consultancy; Acerta: Research Funding; 4D: Research Funding; Eastern Cooperative Oncology Group: Research Funding; Nektar: Research Funding; Sanofi: Research Funding; Morphosys: Research Funding; AbbVie: Consultancy, Research Funding; Bayer: Consultancy, Research Funding; Gilead: Consultancy, Research Funding; Genentech/Roche: Consultancy, Research Funding; Genmab: Consultancy; Novartis: Research Funding; Pfizer: Research Funding; Janssen: Research Funding; National Cancer Institute: Research Funding; Allogene: Research Funding; Amgen: Research Funding; Takeda: Research Funding; Cellectis: Research Funding; Cancer Prevention and Research Institute of Texas: CPRIT Scholar in Cancer Research: Research Funding; BeiGene: Consultancy; Xencor: Research Funding; TG Therapeutics: Research Funding; EMD: Research Funding; Burroughs Wellcome Fund: Research Funding; Ziopharm: Research Funding; Pharmacyclics: Research Funding.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2917-2917
Author(s):  
John Radford ◽  
Ellie White ◽  
Felipe A. Castro ◽  
Anshuman Chaturvedi ◽  
Nathalie Spielewoy ◽  
...  

Introduction: First-line (1L) therapy for diffuse large B-cell lymphoma (DLBCL) can cure ~60% of patients (pts), but ~10-15% do not respond and 20-25% relapse. Salvage chemotherapy with autologous stem cell transplant (ASCT) is standard for 'fit' pts with relapsed or refractory (R/R) disease. However ~50% of pts are ineligible for transplant and most who undergo ASCT will relapse; for these pts options are extremely limited. Here we evaluated R/R DLBCL treatment patterns and outcomes for pts managed at a single UK center to create a detailed 'real-world' comparator for novel therapies. Methods: A detailed retrospective analysis of medical records was undertaken for pts with DLBCL 2006-2017 and a R/R event 2011-2017. Additional eligibility criteria were: age ≥18 years; ≥1 prior anti-CD20 antibody-containing chemo-immunotherapy regimen; no history of high-grade transformation; and no lymphomatous CNS involvement. Pt characteristics, treatments, responses, and overall survival (OS) were reported by line (L) of therapy (2L, 3L, 4L+) in all pts and in refractory pts (defined as no response to or relapse within 6 months of last treatment). Results: Of 2025 pts diagnosed with DLBCL 2006-2017, 89 fulfilled eligibility including a R/R event 2011-2017: 89, 63, and 41 received 2L, 3L, and 4L+ treatment. Median age at 2L was 66 years (range 58-72). 58.4% (n=52) were male and 64.0% (n=57) were stage III/IV; 49.4% (n=44) were ABC subtype and 29.2% (n=26) were GCB. Systemic 2L therapies (≥5% incidence) included R-DHAP (20.2%; n=18), R-GDP (20.2%; n=18), DHAP (10.1%; n=9), R-GCVP (7.9%; n=7), and gemcitabine (5.6%; n=5). In 2L, 23.6% (n=21) of pts underwent ASCT. With each line, regimens became more diverse, with increased use of experimental therapies. Overall response rate was 46.1% in 2L, 27.0% in 3L, and 9.8% in 4L+. In refractory pts, it was 34.8%, 21.2%, and 7.9% (Table). OS is shown in the Figure and Table; 2-year OS was 30.6% in all R/R pts and 20.5% in refractory pts. Median OS was reduced in pts with low ECOG performance status or high LDH. Two-year OS was 71.4% in transplanted pts (n=23) and 16.3% in non-transplanted pts. Conclusions: Despite multiple treatment options, pts with R/R DLBCL have a very poor prognosis, highlighting the need for rapid introduction of more effective therapies. This can be facilitated by robust data such as these, which may be used for comparing outcomes of novel therapies with those expected at a given line of treatment in the 'real world', unrestricted by the requirements of a trial protocol. Results will be compared with those from the SCHOLAR-1 study (Crump et al. Blood 2017;130:1800-8) to provide greater clarity regarding R/R DLBCL treatment outcomes in the 'real world' setting. Disclosures Radford: Seattle Genetics: Consultancy, Honoraria; Takeda: Consultancy, Honoraria, Research Funding; Novartis: Consultancy, Honoraria; ADC Therapeutics: Consultancy, Research Funding; AstraZeneca: Equity Ownership, Research Funding; GSK: Equity Ownership; BMS: Consultancy, Honoraria. Castro:F. Hoffmann-La Roche Ltd: Employment. Chaturvedi:Christie Hospital NHS Trust: Employment. Spielewoy:F. Hoffmann-La Roche Ltd: Employment. Gibb:Takeda: Research Funding. Surinach:Genesis Research: Employment. Shang:F.Hoffmann-La Roche AG: Employment. Wenger:F. Hoffmann-La Roche Ltd: Employment, Equity Ownership. OffLabel Disclosure: "Atezolizumab (atezo) is a programmed death-ligand 1 (PD-L1) blocking antibody. In the United States, atezo is approved for treatment of pts with locally advanced or metastatic urothelial carcinoma who are: not eligible for cisplatin-containing chemotherapy (chemo) and whose tumors express PDL-1, or are not eligible for any platinum-containing chemo regardless of PD L1 status; or have disease progression during or following any platinum-containing chemo, or within 12 months of neoadjuvant or adjuvant chemo. Atezo is also approved: in combination with bevacizumab, paclitaxel and carboplatin for first-line treatment of pts with metastatic non-squamous non-small-cell lung carcinoma (NSCLC) with no EGFR or ALK genomic tumor aberrations, and for pts with metastatic NSCLC who have disease progression during or following platinum-containing chemo; in combination with paclitaxel protein-bound for the treatment of adults with unresectable locally advanced or metastatic triple-negative breast cancer whose tumors express PD-L1; and in combination with carboplatin and etoposide, for the first-line treatment of adults with extensive-stage small cell lung cancer. Atezo is not approved for treatment of pts with multiple myeloma."


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 1-2
Author(s):  
Eva González-Barca ◽  
Johannes Duell ◽  
Federica Cavallo ◽  
Juan-Manuel Sancho ◽  
Zsolt Nagy ◽  
...  

Introduction Patients with diffuse large B-cell lymphoma (DLBCL) that is refractory to primary immunochemotherapy are well recognized as a high-risk population with poor prognosis, and have a typical median overall survival (OS) of 6-13 months [Farooq U, et al. 2017]. Patients with 'double-hit' (MYCand eitherBCL2orBCL6) and 'triple-hit' (MYC, BCL2andBCL6) genetic aberrations are also considered to be a high-risk/poor prognosis group [Davies A. 2019]. Tafasitamab (MOR208) is an Fc-enhanced, humanized, monoclonal anti-CD19 antibody that is under investigation in combination with lenalidomide (LEN) in autologous stem cell transplant (ASCT)-ineligible patients with relapsed/refractory (R/R) DLBCL in the Phase II L-MIND study (NCT02399085) [Salles G, et al. 2020]. We report efficacy data for patients with high-risk DLBCL (primary refractory disease and double/triple-hit lymphoma [DHL/THL]) who received tafasitamab + LEN in L-MIND (data cut-off: Nov 30, 2019; median follow-up for OS, 31.8 months). Methods In the L-MIND study, patients enrolled were aged ≥18 years with R/R DLBCL (1-3 prior systemic therapies, including ≥1 CD20-targeting regimen), with an Eastern Cooperative Oncology Group performance status of 0-2 and ineligible for ASCT. Patients received 28-day cycles of tafasitamab (12 mg/kg intravenously), once weekly during Cycles 1-3 with a loading dose on Cycle 1 Day 4, then every 2 weeks during Cycles 4-12. LEN (25 mg orally) was administered on Days 1-21 of Cycles 1-12. After Cycle 12, progression-free patients received tafasitamab every 2 weeks until disease progression. The primary endpoint was objective response rate (ORR) (partial response [PR] + complete response [CR]), assessed centrally by an independent review committee. Primary refractory disease was defined as no response (CR or PR) to or progression during or within 6 months of frontline DLBCL therapy.MYC,BCL2andBCL6aberrations were determined via fluorescencein situhybridization using tumor biopsy. Results The L-MIND cohort included 15 patients with primary refractory DLBCL and two patients with DHL/THL. Patients with primary refractory DLBCL at baseline had a median age of 73 years (range 48-82; n=9 ≥70 years) and were previously exposed to a median of 2 lines of treatment (range 1-4). Of these patients, ten had stage III/IV disease, ten showed lactate dehydrogenase greater than the upper normal limit, 12 patients had germinal center B-cell DLBCL and eight exhibited intermediate-high or high-risk International Prognostic Index (IPI) status at study baseline. All 15 patients received R-CHOP or equivalent as first-line therapy; two patients previously achieved no response, whereas 13 patients had relapsed within 6 months (ten had achieved a CR and 3 a PR) after frontline therapy. Six patients had received only 1 prior therapy, whereas nine patients had ≥2 lines before L-MIND enrollment. Median time to progression after first-line therapy was 162 days (range 28-182 days); two of 15 patients had progressed within 90 days. Of the 15 patients, 13 were refractory to their last line of therapy before L-MIND. In the 15 patients with primary refractory disease, ORR was 53.3% (95% confidence interval [CI]: 26.6-78.7) and the CR rate was 33.3%, with a 30-month duration of response (DOR) rate of 50% (95% CI: 15.2-77.5) - median DOR not reached (NR). Individual response duration is shown in Figure 1 (swimmer plot): four patients who achieved CR remain in remission after &gt;30 months. Median progression-free survival (PFS) was 5.3 months (95% CI: 0.9-NR) and PFS rate at 30 months was 33.9% (95% CI: 11.0-58.8). Median OS was 13.8 months (95% CI: 1.3-NR) and OS at 36 months was 38.1% (95% CI: 14.6-61.6). Regarding patients with DHL/THL: one with DHL achieved PR only; another patient with THL (also part of the primary refractory subgroup) achieved CR and remains in remission after &gt;30 months. Conclusions The combination of tafasitamab + LEN showed encouraging activity, with a clinically meaningful ORR and CR rate in patients with primary refractory DLBCL, and positive responses in DHL and THL. Patients with primary refractory disease were frequently ≥70 years with stage III/IV disease and poor-risk IPI scores. Although these data should be interpreted with caution due to the small patient subgroup sizes, these clinically relevant results warrant further research with this immunotherapy in patients with difficult-to-treat DLBCL. Disclosures González-Barca: MorphoSys:Other;Janssen:Consultancy, Honoraria;Sandoz:Consultancy;Gilead:Consultancy;Roche:Honoraria;Takeda:Honoraria;Abbvie:Honoraria;Celgene:Consultancy;Kiowa:Consultancy;Celtrion:Consultancy.Duell:Morphosys:Research Funding.Sancho:Bristol-Myers Squibb:Honoraria;Celgene:Consultancy, Honoraria;Gilead:Consultancy, Honoraria;Janssen:Consultancy, Honoraria;Kern-Pharma:Consultancy, Honoraria;Novartis:Consultancy, Honoraria;Roche:Consultancy, Honoraria;Takeda:Honoraria;Celltrion:Consultancy;Sandoz:Consultancy.Nagy:MorphoSys AG:Patents & Royalties.Abrisqueta:Celgene:Consultancy, Honoraria;AbbVie:Consultancy, Honoraria, Speakers Bureau;Roche:Consultancy, Honoraria, Speakers Bureau;Janssen:Consultancy, Honoraria, Speakers Bureau.Panizo:Clínica Universidad de Navarra:Current Employment;Bristol-Myers Squibb, Kyowa Kirin:Speakers Bureau;Janssen, Roche:Membership on an entity's Board of Directors or advisory committees.Augustin:Morphosys:Research Funding;AstraZeneca:Consultancy, Research Funding;Roche:Consultancy;Novartis:Consultancy, Research Funding;Merck:Consultancy;IPSEN:Consultancy, Research Funding;Pfizer:Consultancy, Research Funding;BMS:Consultancy, Research Funding.Weirather:MorphoSys AG:Current Employment.Ambarkhane:MorphoSys AG:Current Employment.Maddocks:Seattle Genetics:Consultancy, Honoraria;Celgene:Consultancy, Honoraria;Pharmacyclics:Consultancy, Honoraria;Morphosys:Consultancy, Honoraria;ADC Therapeutics, AstraZeneca:Consultancy;BMS:Consultancy, Research Funding;Karyopharm:Consultancy.Kalakonda:Celgene:Research Funding.Salles:BMS/Celgene:Honoraria, Other: consultancy or advisory role;Takeda:Honoraria;Karyopharm:Honoraria;Genmab:Honoraria, Other;Debiopharm:Consultancy, Honoraria, Other: consultancy or advisory role;Autolos:Other: consultancy or advisory role;Abbvie:Other: consultancy or advisory role;Roche:Honoraria, Other: consultancy or advisory role;Novartis:Honoraria, Other: consultancy or advisory role;MorphoSys:Honoraria, Other: consultancy or advisory role;Janssen:Honoraria, Other: consultancy or advisory role;Epizyme:Honoraria, Other: consultancy or advisory role;Kite, a Gilead Company:Honoraria, Other: consultancy or advisory role .


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 213-213
Author(s):  
Zijun Y. Xu-Monette ◽  
Bouthaina S. Dabaja ◽  
Alexander Tzankov ◽  
Carlo Visco ◽  
Roberto N. Miranda ◽  
...  

Abstract Context and objective Approximately 5-10% of diffuse large B-cell lymphoma (DLBCL) patients carry MYC gene translocations (MYC-translocation+) with a poor prognosis after standard chemotherapy. MYC-translocation+ DLBCL patients carrying BCL2 translocations (MYC+/BCL2+ double-hit lymphoma) have a worse survival. The efficacy of adjuvant radiotherapy in this setting is unknown. The purpose of this study is to evaluate the efficacy of radiotherapy as a part of the therapeutic regimen for patients with MYC-translocation+ DLBCL. Patients and methods From the International DLBCL R-CHOP consortium program, we selected 581 patients with de novo DLBCL treated with standard R-CHOP immunochemotherapy (diagnosed and treated from 2000 to 2010). We excluded patients with transformed DLBCL, primary mediastinal, cutaneous, testicular or central nervous system large B-cell lymphomas, patients with HIV infection, and patients not treated with R-CHOP. The median follow-up was 54.9 months. Fluorescence in situ hybridization assessing MYC was performed for all the patients (n=581) and results were correlated with available clinical data to identify clinicopathologic features associated with MYC translocation, and to evaluate the prognostic significance of MYC translocations regarding overall survival (OS, from the time of diagnosis to death from any cause) and progression-free survival (PFS, from the time of diagnosis to relapse or death from any cause). In the MYC-translocation+ DLBCL group, 38 patients received chemotherapy alone and 21 patients received chemotherapy with adjuvant radiation therapy. The clinicopathologic features and survival of MYC-translocation+ DLBCL patients treated with (n=21) and without radiotherapy (n=38) after immunochemotherapy were compared to in order to evaluate the radiotherapy efficacy and other confounding factors. Results MYC translocations were detected in 59 DLBCL patients (10.2%). Patients with MYC-translocation+ DLBCL more often had bulky tumors, involvement of multiple extranodal sites, and poorer OS (hazard ratio [HR]: 2.0, 95% confidence interval [CI]: 1.20 - 3.35, P= .0083) and PFS (HR: 1.96, 95% CI: 1.22 - 3.13, P= .0005) independent of the International Prognostic Index score. Poor survival was primarily attributable to patients with MYC+/BCL2+ double-hit DLBCL who were predominantly of germinal center B-cell subtype. Among MYC-translocation+ DLBCL patients, a better survival was achieved in patients who received radiotherapy (for OS, HR: .32, 95% CI: .15 - .71, P= .0049; for PFS, HR: .35, 95% CI: .17 - .73, P= .0043). Conversely, radiotherapy abolished the adverse impact of MYC translocations. In addition, radiotherapy was associated with better survival in the subset of patients with MYC+/BCL2+ double-hit lymphoma (P = .017 for OS, and P = .05 for PFS). However, owing to the common use of radiotherapy as consolidation therapy, the favorable prognoses in the group of patients who received radiotherapy could also be attributed to limited-stage disease and more frequent complete remission (CR) after first-line treatment and therefore these factors confound interpretation of the data. To address these issues, we evaluated radiotherapy efficacy in separate patient groups: patients who achieved CR, non-CR (PR/SD/PD) patients, and patient with stage I/II, or stage III/IV disease. The efficacy of radiotherapy appeared more apparent in patients with advanced disease who did not achieve CR after first-line chemotherapy. Multivariate analysis after adjustment for stage and IPI score validated that radiotherapy significantly improved OS (HR: .28, 95% CI: .10 - .81, P= .018) and PFS (HR: .32, 95% CI: .13 - .80, P= .015) of MYC-translocation+ DLBCL patients. Conclusions The presence of MYC translocations in DLBCL is an important biomarker that facilitates prognostic prediction and treatment stratification independent of the IPI score. For chemoresistant MYC-translocation+ DLBCL patients, radiotherapy seems to be an effective adjuvant regimen likely due to the higher frequency of extranodal involvement and bulky disease in MYC-translocation+ DLBCL patients. Our results provide a rationale for larger scale studies to assess the potential role of radiotherapy in the management of MYC-translocation+ DLBCL patients, particularly patients with MYC+/BCL2+double-hit DLBCL. Disclosures: Winter: Millenium: Research Funding; Novartis : Research Funding; Pfizer (Wyeth): Research Funding; Seattle Genetics: Research Funding; Spectrum: Research Funding; Janssen (Pharmacyclics): Research Funding; Spectrum (Allos): Consultancy; Sanofi Aventis: Consultancy; Tgen: Consultancy; AMBIT Biosciences (Spouse): Research Funding; Celgene (Spouse): DSMB, DSMB Other, Research Funding; Ariad Pharmaceuticals (Spouse): Research Funding; Novartis (Spouse): Consultancy, Research Funding; Amgen (Spouse): Consultancy, Research Funding; Astellas (Spouse): Research Funding; Caremark/CVS: Consultancy; Pfizer (Spouse): Consultancy; Sanofi Aventis (Spouse): DSMB, DSMB Other; Bristol Myers Squibb (Spouse): DSMB, DSMB Other; UptoDate, Inc.(Spouse): Patents & Royalties.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3577-3577
Author(s):  
Sandra Kanan ◽  
Ajay K Gopal ◽  
Ryan D Cassaday ◽  
Andrei R. Shustov ◽  
Brian G. Till ◽  
...  

Abstract Background : Increasing data from single-arm studies suggest da-EPOCH-R may be optimal therapy for subsets of aggressive B-NHL, including primary mediastinal B-cell lymphoma, germinal-center DLBCL, and forms with MYC dysregulation. However, da-EPOCH-R administration is considerably more complex than R-CHOP, requiring twice-weekly laboratory monitoring and critical dose adjustments. Major studies in DLBCL (Wilson Blood 2002, Wilson JCO 2008) employ daily GCSF in particular. When daily GCSF is used, a mean overall dose intensity equivalent to dose level 2 is achieved. In younger patients with mediastinal B-cell lymphoma, more than half of patients attained dose level 4. We hypothesized that the use of da-EPOCH in general would have increased over the last decade. In a key subset of patients receiving first-line da-EPOCH-R for aggressive B-NHL, we sought to identify frequency of peg-filgrastim use, as a fundamental deviation from protocol-specified therapy, and identify maximum dose level achieved with peg-GCSF vs. daily GCSF. Given their similarity, we did not anticipate that choice of growth factor would predict likelihood of reaching dose level 4. Methods: We identified all patients receiving da-EPOCH from 2005 to 2015 at the University of Washington and Seattle Cancer Care Alliance and analyzed cases for baseline features, growth factor employed, and maximum dose level attained. Patients with DLBCL and variants (PMBCL, transformed lymphoma, DLBCL-PTLD) and BCL-unclassifiable (BCLU), who received at least 4 cycles of da-EPOCH with rituximab as first-line standard of care therapy (not in context of a clinical trial) were subject to detailed analysis regarding growth factor and dose level achieved. IRB approval was obtained. Results: 165 patients receiving da-EPOCH were initially identified, demonstrating an over 5-fold increase in use of this regimen over the 10-year period (Figure 1). Of these, 73 patients with DLBCL and BCLU met the above criteria, receiving da-EPOCH-R as first-line therapy. Median age was 60 (range 24-78) and patients received a median of 6 cycles (range 4-8; 75% of pts received 6 cycles) of da-EPOCH-R. Most patients (44/73, or 60%) received peg-GCSF rather than daily GCSF with da-EPOCH-R. Overall, the median, highest dose level during first-line therapy was 2 without a difference in groups receiving peg-GCSF or daily GCSF. 61% of patients attained dose level of 2, 41% achieved level 3, and 15% achieved level 4. The proportion of patients who achieved dose level 4 was comparable in the peg-GCSF group (11%) and daily GCSF (21%, p=.24, chi-2). Conclusions: Infusional da-EPOCH is being increasingly used, despite limited single arm data supporting its benefit. For first-line therapy of aggressive B-NHL, da-EPOCH-R is being broadly applied to older subgroups of patients. In that subgroup, peg-GCSF is used more than half of the time. In our population, only 15% achieved a dose level of 4 or higher, compared to over half of patients with PMBCL and a median age of 30 (Dunleavy NEJM 2013). Age is well known as a predictor of da-EPOCH dose intensity (Wilson 2002). Variations from the published protocol due to clinical judgment, as well as patient factors, are also possible factors influencing our dosing findings. Peg-GCSF vs. GCSF did not appear to impact the ability to attain dose level 4, although our study design cannot answer this definitely. Since peg-GCSF is cost effective compared to daily GCSF in lymphoma treated with CHOP (Lyman Curr Med Res Opin 2009) and more convenient, our data suggests that peg-GCSF may be a reasonable strategy to support the da-EPOCH-R regimen. Disclosures Gopal: Seattle Genetics: Research Funding. Shadman:Gilead: Honoraria, Research Funding; Acerta: Research Funding; Pharmacyclics: Honoraria, Research Funding; Emergent: Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5040-5040
Author(s):  
Joanna Romejko-Jarosinska ◽  
Michal Osowiecki ◽  
Beata Ostrowska ◽  
Martyna Kotarska ◽  
Katarzyna Domanska-Czyz ◽  
...  

Abstract Background: Secondary central nervous system (CNS) involvement is an unfavorable risk factor with an impact on overall survival (OS) in malignant lymphoma. Methods: We conducted a retrospective analysis of secondary CNS involvement in patients diagnosed with Diffuse Large B-Cell lymphoma (DLBCL) on the first line treatment and after the end of treatment. We collected data on 361 patients diagnosed with DLBCL. Results: The median age (range) was 66 (17-91) years. All patients received first-line R-CHOP21 (rituximab, cyclophosphamide, vincristine, doxorubicin, prednisone) between 2006-2011. Patients with primary involvement of testis, nasopharynx, orbit and CNS received CNS prophylaxis with 15 mg methotrexate intrathecally (IT). Secondary CNS involvement was confirmed in 18 (4.9%) patients which stands for 24.6% (18/73) patients with relapsed lymphoma. Median time from first R-CHOP to lymphoma relapse was 10 months (range 3-33). CNS involvement in first 12 months of treatment was 3.9% (95% CI: 1.8%, 5.8%). Median (range) OS from diagnosis of CNS involvement was 3 (3-79) months. Clinical stage III/IV, IPI 4, breast, 2 or more extranodal sites and NCCN-IPI unfavorable site involvement increased the risk of CNS involvement. Three patients who had received methotrexate IT prophylaxis had CNS involvement at relapse. No patient with primary testis involvement had CNS relapse. Conclusion: Less than 5% of patients had secondary CNS involvement on first line treatment and after end of treatment. Advanced stage, high-risk IPI, 2 or more extranodal sites of involvement increased the risk of CNS involvement after first-line treatment for DLBCL. CNS prophylaxis appears effective in patients with primary testicular lymphoma Disclosures Walewski: Karyopharm: Consultancy; Roche: Consultancy, Honoraria, Other: travel, accommodation, Research Funding; Celgene: Honoraria, Other: travel, accommodation, Research Funding; Gilead: Consultancy, Honoraria, Other: travel, accommodation; Takeda: Consultancy, Honoraria, Other; Ariad: Consultancy; Janssen-Cilag: Consultancy; Mundipharma: Consultancy, Honoraria, Research Funding; Genetics: Other: travel, accommodation; Teva: Consultancy, Honoraria; Seattle: Other: travel, accommodation; GSK/Novartis: Research Funding; Boehringer Ingelheim: Consultancy; Sanofi: Honoraria, Other: travel, accommodation; Servier: Consultancy.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1686-1686
Author(s):  
Stephen D. Smith ◽  
Ryan C. Lynch ◽  
Brian G. Till ◽  
Andrew J. Cowan ◽  
Qian V. Wu ◽  
...  

Abstract Background: Rituximab with cyclophosphamide, doxorubicin, vincristine, and prednisone (RCHOP) remains standard therapy for previously untreated diffuse large B-cell lymphoma (DLBCL). However, a substantial proportion of patients (pts) will relapse. The anti-PD-1 monoclonal antibody pembrolizumab has shown modest efficacy in previously treated DLBCL. We postulated that the first-line setting, with relatively intact host immunity and coexistence of malignant cells with T-cells in the microenvironment, may represent an opportunity for effective immune checkpoint inhibition. We carried out the first known prospective trial of anti-PD1 therapy with anthracycline chemotherapy in lymphoma, to evaluate the safety of this combination. Methods: Pts age 18 years or older with previously untreated DLBCL, transformed lymphoma and grade 3B follicular lymphoma with a plan for 6-cycle, curative-intent RCHOP were eligible. Steroids within 7 days of treatment, active autoimmune disease, or history of pneumonitis were excluded. This phase I study combined pembrolizumab (200 mg IV q 3 weeks x 6) to RCHOP x 6, with supportive care per standard practice. A 30-pt sample size permitted estimation of the rate of clinically relevant grade 3-5 toxicity, assumed to occur in 40% of pts with RCHOP alone. A stopping rule for more than 3 instances of non-relapse death or inability to complete 6 cycles of RCHOP for any reason was applied. Secondary endpoints included response rates, overall (OS) and progression-free survival (PFS). Baseline PDL1 expression was analyzed centrally (Merck/Qualtek). Peripheral blood flow cytometry for changes in PD1, PDL-1, and PDL-2 subsets were performed. After PET-based response assessment, pts were followed for relapse and survival. Results: Since March 2016, 29 pts were treated on study; 28 have finished all therapy. One is currently on therapy, and 1 remains to be enrolled. Pt characteristics are in Table 1. In 29 pts to date, 18 grade 3-5 clinically significant AE's occurred in 13 unique pts (13/29, 45%). 16 SAE's occurred in 11 pts (11/29, 38%); none qualified as unanticipated events. Two deaths occurred: One in the first accured patient, who had extensive gastric involvement by DLBCL, and died during cycle 1 of bleeding from the responding tumor bed despite maximal inpatient intervention. The other death occurred due to steroid-refractory GVHD after haploidenticial allogeneic transplant for relapsed disease. Four immune-related adverse events (IRAE) occurred: grade 3 rash, resolving with oral steroids and not recurring with further pembrolizumab, grade 1 hyperthyroidism, grade 2 colitis, and 1 episode of grade 3 pneumonitis. This case of pneumonitis was the only immune-related SAE, occurring in a 78 yo with a history of tobacco use and COPD, and resulted in stopping therapy after cycle 3. Among 27 completing treatment (excluding the pt who died during cycle 1), average anthracycline dose was 95% of expected; anthracycline relative dose intensity (RDI: delivered dose intensity/standard dose intensity) was 94%. PET response assessment among 26 evaluable pts showed 18 CR (69%), 7 PR, and 1 primary refractory disease. Among 7 PR, 1 relapsed, 3 had negative biopsies, and 3 remain in remission. Median follow-up of survivors is 13 months and 2 relapses (1 primary progressive disease, 1 after PET PR) occurred. One-year PFS is 87% (Figure 1). Baseline PDL-1 staining available for 19 pts showed a median % tumor cell staining of 30% (moderate 2+ plus strong 3+ staining; range, 0-100%). Tumor PDL1 was ≤5% in 4, 10-29% in 5, 30-49% in 4, and ≥50% in 6 pts. Both EBV+ DLBCL had 60% tumor PDL1 expression. 1 relapsing pt had no detectable tumor PDL1, and the THRLBL failed for technical reasons. Conclusions: RCHOP with pembrolizumab 200 mg q 3 weeks x 6 cycles is safe, with toxicity similar to RCHOP alone and only 4 IRAE's. Anthracycline dose intensity, CR rate, and PFS are favorable. PDL1 tumor cell expression of 10% or more was observed in 15/19 pts. These data support further study of pembrolizumab with chemotherapy in untreated lymphoid malignancies. Final response/progression, correlative studies, and survival data will be presented in December after final accrual. Disclosures Smith: Merck Sharp and Dohme Corp.: Consultancy, Research Funding; Pharmacyclics: Research Funding; Portola Pharmaceuticals: Research Funding; Acerta Pharma BV: Research Funding; Genentech: Research Funding; Seattle Genetics: Research Funding; Incyte Corporation: Research Funding. Lynch:Johnson Graffe Keay Moniz & Wick LLP: Consultancy; Incyte Corporation: Research Funding; Takeda Pharmaceuticals: Research Funding; T.G. Therapeutics: Research Funding; Rhizen Pharmaceuticals S.A.: Research Funding. Till:Mustang Bio: Patents & Royalties, Research Funding. Cowan:Sanofi: Research Funding; Juno Therapeutics: Research Funding; Abbvie: Research Funding; Janssen: Research Funding. Shadman:Acerta Pharma: Research Funding; Genentech: Research Funding; Verastem: Consultancy; Gilead Sciences: Research Funding; Qilu Puget Sound Biotherapeutics: Consultancy; Celgene: Research Funding; Genentech: Consultancy; Beigene: Research Funding; Pharmacyclics: Research Funding; AbbVie: Consultancy; Mustang Biopharma: Research Funding; TG Therapeutics: Research Funding; AstraZeneca: Consultancy. Shustov:Seattle Genetics: Research Funding. Cassaday:Amgen: Consultancy, Research Funding; Incyte: Research Funding; Seattle Genetics: Other: Spouse Employment, Research Funding; Merck: Research Funding; Jazz Pharmaceuticals: Consultancy; Adaptive Biotechnologies: Consultancy; Pfizer: Consultancy, Research Funding; Kite Pharma: Research Funding. Gopal:Pfizer: Research Funding; Janssen: Consultancy, Research Funding; BMS: Research Funding; Takeda: Research Funding; Incyte: Consultancy; Spectrum: Research Funding; Gilead: Consultancy, Research Funding; Merck: Research Funding; Aptevo: Consultancy; Asana: Consultancy; Seattle Genetics: Consultancy, Research Funding; Brim: Consultancy; Teva: Research Funding.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 10-11
Author(s):  
Danny Luan ◽  
Yiyuan Wu ◽  
Jordan S. Goldstein ◽  
Sarah C. Rutherford ◽  
John P. Leonard ◽  
...  

Background: Positron emission tomography-computed tomography (PET-CT) has become the primary modality for initial staging in diffuse large B-cell lymphoma (DLBCL) [Barrington et al, J Clin Oncol 2014; Cheson et al, J Clin Oncol 2014]. Recently, the role of a staging bone marrow biopsy (BMB) has become less clear. A meta-analysis suggested that PET-CT can miss bone marrow involvement [Adams et al, Eur J Nucl Med Mol Imaging 2014], but recent guidelines on staging suggested that BMB should be optional. We analyzed temporal trends in use of different screening modalities and evaluated the association between patient outcomes and staging modality (PET-CT with BMB compared to either PET-CT without BMB or CT with BMB) in patients with DLBCL in SEER-Medicare. We hypothesized that use of PET-CT as screening would increase over time and that use of BMB would decrease over time. We also hypothesized that use of BMB would not be associated with an improvement in overall survival in patients staged with PET-CT. Methods: 70,858 patients diagnosed with DLBCL in SEER-Medicare were identified with ICD-O-3 SEER histology codes 9680 and 9684. In this analysis, we included 7,159 patients who were diagnosed between 2002 and 2015, were treated with first-line rituximab, cyclophosphamide, vincristine, and doxorubicin chemotherapy regimens, and had claims for PET-CT and/or BMB within 30 days on either side of diagnosis. The patients were divided into screening categories of patients receiving PET-CT without BMB (PET-CT w/o BMB), patients receiving BMB with CT (i.e., no PET-CT) (CT w/ BMB), and patients receiving both screening modalities (PET-CT w/ BMB). Overall survival (OS) was calculated from date of diagnosis to death. Kaplan-Meier (KM) curves were used to estimate survival probabilities across screening categories and log-rank tests (LRT) were used to statistically evaluate differences between OS curves. Cox proportional hazards (CoxPH) models were used to estimate hazard ratios prior to and following adjustment for confounders. Results: 2,059 patients received PET-CT w/o BMB, 1,539 received CT w/ BMB, and 3,561 received PET-CT w/ BMB. Proportions of patients receiving PET-CT w/ BMB and PET-CT w/o BMB increased across years of diagnosis (36.7 to 51.9% and 17.4 to 36.1%, respectively, between 2002-2005 and 2013-2015), while the proportion of patients receiving CT w/ BMB decreased across years of diagnosis (45.8 to 12.0%) (Table 1). Similarly, the proportion of patients receiving BMB independent of imaging modality also decreased (28.8% to 16.8%). In bivariate analyses, categories of screening were significantly associated with age category, sex, year of diagnosis, race, stage, nodal status, poor performance status, Charlson Comorbidity Index (CCI), number of first-line cycles, and geographic classification, with receipt of PET-CT w/ BMB being more common in patients who are younger, male, diagnosed in later years in urban settings, and received &gt;4 first-line cycles, with earlier stage at diagnosis, nodal disease, good performance status, and lower CCI (Table 1). In unadjusted KM analysis, median survival among patients receiving PET-CT w/ BMB was 8.7 years, compared to 8.1 and 7.6 years among patients receiving PET-CT w/o BMB and CT w/ BMB, respectively (LRT P&lt;0.0001; Figure 1). In fully adjusted CoxPH models, patients receiving PET-CT w/ BMB did not have a significantly better OS compared to those receiving PET-CT w/o BMB (HR: 0.93; 95% CI, 0.84-1.02; P=0.1075). Separately, patients receiving CT w/ BMB had a significantly worse OS compared to those receiving PET-CT w/o BMB (HR: 1.13; 95% CI, 1.02-1.26; P=0.0212), even though OS was not significantly associated with year of diagnosis (P=0.6613). Conclusions: In this SEER-Medicare cohort of 7,159 patients with newly diagnosed DLBCL, we found temporal trends supporting increased use of pretreatment staging PET-CT and decreased use of BMB. We also found that use of PET-CT w/ BMB did not provide a survival benefit as compared to use of PET-CT w/o BMB. These data would suggest that use of BMB could be spared in patients newly diagnosed with DLBCL over the age of 65, unless otherwise clinically indicated. Interestingly, the finding of no OS benefit across years of diagnosis suggests that improvements in outcomes in recent trials may be due to patient selection. Given the limitations associated with registry data, the study warrants replication in more complete DLBCL data sets. Disclosures Rutherford: Genentech/Roche: Research Funding; Karyopharm: Consultancy, Research Funding; Regeneron: Research Funding; Celgene: Consultancy; Kite: Consultancy; Dova: Consultancy; Seattle Genetics: Consultancy; AstraZeneca: Consultancy; LAM Therapeutics: Research Funding; Heron: Consultancy; Juno: Consultancy. Leonard:Miltenyi: Consultancy; Karyopharm: Consultancy; Gilead/Kite: Consultancy; Sutro: Consultancy; Bayer: Consultancy; BMS/Celgene: Consultancy; MEI Pharma: Consultancy; Epizyme: Consultancy; Roche/Genentech: Consultancy; Regeneron: Consultancy; ADC Therapeutics: Consultancy; GenMab: Consultancy; AstraZeneca: Consultancy. Martin:Kite: Consultancy; Morphosys: Consultancy; Regeneron: Consultancy; Karyopharm: Consultancy, Research Funding; Janssen: Consultancy; Sandoz: Consultancy; I-MAB: Consultancy; Bayer: Consultancy; Incyte: Consultancy; Cellectar: Consultancy; Beigene: Consultancy; Celgene: Consultancy; Teneobio: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document