Concordance with National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines (CPGs): A Quality of Care Tool for Newly Diagnosed Patients (pts) with Mantle Cell Lymphoma (MCL).

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3328-3328
Author(s):  
Maria Alma Rodriguez ◽  
Anna Ter Veer ◽  
Auayporn Nademanee ◽  
Joyce Niland ◽  
Eva Lepisto ◽  
...  

Abstract Introduction: In 1999, the NCCN published its first NHL CPG and in 2000, established the NCCN NHL Outcomes Database Project to monitor patterns of care, CPG concordance and outcomes in participating institutions. We report here on clinical characteristics and CPG concordance among newly diagnosed (dx) pts with MCL in the database. Methods: We prospectively collected demographic, staging, and treatment information on consecutive pts with MCL presenting at 5 geographically diverse NCCN institutions (Dana-Farber, Roswell Park, City of Hope, Fox Chase and MD Anderson). We assessed concordance with 2 CPG’s relevant to the concordance impacting the majority of pts: 1) bone marrow biopsy (BMBx) as part of the initial work-up and 2) use of an endorsed first line regimen among pts with stage III/IV disease. CPG concordance was assessed by comparing each pt’s care against the version on the NCCN guideline in effect at the time the pt was diagnosed. Results: Between 7/2000 and 10/2005, we enrolled 132 MC evaluable pts. Median age was 58; 43% had high-intermediate or high risk disease according to the IPI at presentation; 123 (93%) pts presented with stage III/IV disease. The median follow-up was 22.6 months. Overall, 91% of pts underwent a staging BMBx as recommended by the guidelines. Concordance varied by institution, low 78% to high 98%. Among 123 pts with stage III/IV disease, first-line therapy was concordant with CPG recommendations in only 59%. Use of rituximab accounted for 92% of all non-concordance. When the guidelines were modified in 2003 to include rituximab as an option for first-line therapy of MCL, concordance rose from 31% (2000) to 100% (2003–5). Of concordant pts receiving combination chemotherapy, 33% received CHOP-based standard dose therapy and 62% received dose-intense therapy. NCCN guidelines consider all therapy administered as part of a clinical trial to be concordant; trial-directed treatment accounted for 42% of concordant care. Conclusions: Our data suggest that the majority of MCL pts in these centers receive care that is concordant with current standards. In this subgroup of patients, participation in clinical trials occurred at an impressively high rate. However, not all pts undergo BMBx as a routine component of staging as recommended by NCCN guidelines suggesting that this is an area for potential quality improvement. This study also highlights that differences in management exist even within national comprehensive cancer centers. Because long-term follow-up is possible with this database, future studies will assess the initial treatment and guideline concordance on long-term outcomes in this unique group of pts.

2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 15015-15015 ◽  
Author(s):  
J. D. Zubkus ◽  
J. D. Hainsworth ◽  
D. R. Spigel ◽  
J. F. Patton ◽  
D. L. Shipley ◽  
...  

15015 Background: Topotecan (T) is standard therapy for patients (pts) with relapsed platinum refractory ovarian cancer (OC). The addition of T to paclitaxel/carboplatin (PC) may improve the first-line therapy for pts with stage III/IV OC. Methods: The primary endpoints were to assess the toxicity and response rate of TPC in previously untreated pts with advanced OC. Pts with previously untreated stage III/IV OC with performance (PS) 0 or 1, normal hematologic and organ function were eligible. Topotecan 1mg/m2 per day IV days 1, 2, and 3; paclitaxel 175mg/m2 IV on day 3 and carboplatin (AUC=5) IV on day 3 were administered at 21-day intervals for 6 cycles with standard dose modifications for toxicities. Responses were assessed clinically after 2 cycles and at completion of therapy. Second look laporatory (SLL) was required for pts without evaluable tumor. Results: 50 pts were enrolled: age range 27–79 (median 63); performance status 0=16, 1=34; suboptimal debulking surgery (tumor > 1 cm) 32; 70% high grade tumors. Intent to treat response rates: complete response (CR) 20 pts (40%); partial response (PR) 12 pts (24%); stable (S) 13 pts (26%); progression 2 pts (4%); not evaluable 3 pts (6%). 13 pts had SLL with 5 CR, 5 PR and 3 S. Toxicities included: grade 3/4 neutropenia/thrombocytopenia 84%/44%; grade 3/4 fatigue/infection 10%/10%; no treatment-related deaths. Median progression-free survival (PFS) for all pts was 13.6 months and 1-, 2-, 3-year PFS 60%, 42%, 36%. Median survival 33.9 months for all pts with 1-, 2-, 3-year survivals 90%, 65%, 49%. Conclusions: The combination of TPC is active and relatively well tolerated with PC given on day 3. The response rates, PFS, and survivals appear similar to standard regimens. Randomized prospective trials will be required to determine the value of T added to PC for first-line therapy of advanced OC. [Table: see text]


2018 ◽  
Vol 36 (15_suppl) ◽  
pp. 7542-7542 ◽  
Author(s):  
Jonathan W. Friedberg ◽  
Andres Forero-Torres ◽  
Beata Holkova ◽  
Jerome H. Goldschmidt ◽  
Ralph V. Boccia ◽  
...  

2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 9001-9001 ◽  
Author(s):  
Charles M. Rudin ◽  
Mark M. Awad ◽  
Alejandro Navarro ◽  
Maya Gottfried ◽  
Solange Peters ◽  
...  

9001 Background: Pembro monotherapy showed durable antitumor activity as third-line or later therapy for metastatic SCLC, leading to FDA approval in that setting. KEYNOTE-604 was a double-blind, phase 3 study of pembro + EP vs placebo + EP as first-line therapy for ES-SCLC (NCT03066778). Methods: Eligible patients (pts) with previously untreated ES-SCLC and no untreated CNS metastases were randomized 1:1 to pembro 200 mg Q3W or saline placebo for up to 35 cycles plus 4 cycles of standard-dose EP. Pts with CR or PR after cycle 4 could receive PCI at investigator discretion. Randomization was stratified by platinum choice (carboplatin vs cisplatin), ECOG PS (0 vs 1), and LDH (≤ULN vs > ULN). Primary endpoints were OS and PFS (RECIST v1.1, blinded central review) in the ITT population. ORR, DOR, and safety were secondary endpoints. OS and PFS treatment differences were assessed by the stratified log-rank test. The protocol specified 2 interim analyses (IAs) and a final analysis (FA). Prespecified efficacy boundaries were one-sided P = 0.0048 for PFS at IA2 (prespecified final PFS analysis) and 0.0128 for OS at FA. Results: 453 pts were randomized. 223/228 pts assigned to pembro + EP and 222/225 assigned to placebo + EP received ≥1 dose of assigned treatment; 1 pt assigned to pembro + EP received placebo + EP in error. Median age was 65 y, 74% had ECOG PS 1, and 57% had LDH > ULN; more pts in the pembro + EP arm had baseline brain metastases (14% vs 10%). At FA (median follow-up, 21.6 mo), 9% of pts in the pembro + EP arm and 1% in the placebo + EP arm remained on study treatment; 12% and 14% received PCI. At IA2 (median follow-up, 13.5 mo), pembro + EP significantly improved PFS in the ITT population (HR 0.75 [95% CI 0.61-0.91], P = 0.0023; median 4.5 vs 4.3 mo). At FA, pembro + EP prolonged OS in the ITT population, but the significance threshold was not met (HR 0.80 [95% CI 0.64-0.98], P = 0.0164; median 10.8 vs 9.7 mo). In a post hoc analysis of OS in the as-treated population, the nominal P value was smaller than the significance threshold (HR 0.78 [95% CI 0.63-0.97], P = 0.0124). ORR at FA was 71% for pembro + EP vs 62% for placebo + EP; median DOR was 4.2 vs 3.7 mo. Observed AEs were as expected; any-cause AEs were grade 3-4 in 77% vs 75%, grade 5 in 6% vs 5%, and led to discontinuation in 15% vs 6%. Conclusions: Pembro + EP significantly improved PFS and prolonged OS compared with placebo + EP as first-line therapy for pts with ES-SCLC. No unexpected toxicities were seen with pembro + EP. These data support the benefit of pembro-containing regimens for ES-SCLC. Clinical trial information: NCT03066778.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 904-904 ◽  
Author(s):  
Peter Reimer ◽  
Thomas Ruediger ◽  
Tobias Schertlin ◽  
Eva Geissinger ◽  
Florian Weissinger ◽  
...  

Abstract Peripheral T-cell lymphomas (PTCL) represent a heterogeneous group of non-Hodgkin’s lymphomas, which in general show a poor outcome following conventional chemotherapy. Long-term remissions are achieved in only 15 to 35 %. However, the impact of more aggressive therapeutic approaches such as myeloablative therapy with autologous stem cell transplantation (ASCT) as first line therapy is poorly defined mainly due to the lack of prospective PTCL-restricted studies. In 6/00 we initiated the first prospective PTCL-restricted multicenter study of myeloablative radiochemotherapy in primary diagnosed PTCL. The results of the first 30 patients (pts) are in press. We update our data on all pts entering the study. Study design: Pts < 65 years with PTCL of all subtypes without primary cutaneous lymphoma and ALK1 expressing anaplastic large cell lymphoma were included. Treatment consisted of 4–6 courses of CHOP protocol followed by DexaBEAM or ESHAP regimen and collection of stem cells. Subsequently pts underwent total body irradiation (TBI) and high dose cyclophosphamide chemotherapy (60 mg/kg body weight) with ASCT. Patient characteristics: From 6/00 to 8/04 65 pts (42 male) with a median age of 50 years were enrolled. Main subtypes were Peripheral T-cell lymphoma not otherwise specified (NOS, n= 26) and Angioimmunioblastic T-cell lymphoma (AILT, n= 19). According to the Ann Arbor classification, 81% of the pts had stage III/IV disease. The International Prognostic Index (IPI) was low/low intermediate in 54% and intermediate high/high in 46% of the pts, respectively. Results: So far 54 of 65 pts are eligible for evaluation, while the remaining 11 pts are still under therapy. Thirty-three pts could be transplanted (61%). After a median follow up of 10 months after transplantation 22 pts (67%) are in sustained remission and 8 pts (27%) had relapsed. Post transplantation two pts died treatment-related (one secondary AML, one multiorgan failure). Twenty-one pts (39%) did not proceed to ASCT mainly due to progressive disease (n= 16). Treatment-related toxicity was comparable to other high-dose studies in malignant lymphomas. Conclusion: Our data show feasibility and efficacy of first-line ASCT following myeloablative radiochemotherapy in PTCL. Sustaining remission seems achievable for a majority of pts. However, additional treatment strategies are required to prevent early progression prior myeloablative therapy. Longer follow-up is necessary to confirm long-term remission rate.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1542-1542
Author(s):  
Kristen Gonter-Aubin ◽  
Daniel Alan Kerr ◽  
Ashley Rose ◽  
Thanh Lana Nguyen ◽  
Julio C. Chavez ◽  
...  

Background: Hairy cell leukemia (HCL) is an indolent mature B-cell disorder that comprises ~2% of all leukemias. Purine nucleoside analogues (PNA) such as cladribine and pentostatin remain the standard initial treatment with high response rate, however frequent late relapses occur. The addition of rituximab (R) has shown efficacy as a single agent and in combination in the first- and second-line settings . Recent data suggests that the addition of R to PNA increased the minimal residual disease (MRD) negative (neg) and duration of response (DOR) when compared to PNA alone. In this study, we examined the impact of rituximab (R) in the long term outcomes of HCL from a single institution experience. Methods: An IRB-approved retrospective analysis was performed including patients with HCL or HCL-variant who were seen at Moffitt Cancer Center between 1/2000 and 2/ 2019. Primary endpoints were overall (OS) and progression-free survival (PFS). Secondary endpoints included overall response rate (ORR). Complete Remission (CR) was determined based off of accepted criteria of Hgb &gt; 11gr/dL, platelets &gt; 100,000/µL and ANC &gt; 1500/µL. Statistical comparisons and survival analyses were performed using SPSSTM Statistics and GraphPad PrismTM. Results: We identified 124 cases with available treatment data and follow up (82 HCL, 15 CD25-negative HCLv). The median follow-up was 72 months. The median age was 57 years (27 - 89+). The 5 and 10 year OS was 97% and 91%, respectively, with 2 deaths reported secondary to HCL or complications from HCL. The median PFS for all patients receiving first-line therapy was 92 months. Clinical risk factors present at diagnosis that were independently associated with worse PFS after first-line therapy were weight loss (HR 2.27, p = 0.057), recurrent infections (HR 2.50, p=0.009) and splenomegaly (HR 1.87, p=0.047),, hemoglobin less than 11 gr/dL (HR 3.12, p=0.016), and negative CD25 (HR 2.14, p=0.055). In multivariate analyses, negative CD25, recurrent infections and Hgb &lt; 11 gr/dL were associated with worse PFS after first line therapy (p = 0.002). Most patients received PNA alone or PNA+R (96 and 18, respectively) as first line with a higher CR rate in the PNA+R group (100% vs 71%) and a higher median PFS for [not reached (NR) and 82 months, respectively, p = 0.165] with 2 relapses in the PNA+R group (both at 30 months). In the 2nd line setting, PNA+R (n = 11) was also associated with a higher CR rate than PNA alone (n = 22, 82% vs 64%) with only 1 relapse in the PNA+R group at 97 months. The median PFS for 2nd line PNA+R vs 2-CdA was 97 and 43 months, respectively (p = 0.253). Conclusions: In this study, we report a large, retrospective, single-institution dataset of HCL and HCLv patients with long-term follow-up. The addition of R may improve the PFS in patients with HCL in the first and 2nd -line settings however a longer follow up is needed. A recently described prospective randomized trial in the use of concomitant rituximab plus PNA has reported a higher MRD free CR in comparison with PNA alone but not PFS yet. Disclosures Chavez: Celgene: Membership on an entity's Board of Directors or advisory committees; Karyopharm: Membership on an entity's Board of Directors or advisory committees; Bayer: Membership on an entity's Board of Directors or advisory committees; Genentech: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Kite: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Novartis: Membership on an entity's Board of Directors or advisory committees. Pinilla Ibarz:Novartis: Consultancy; Bristol-Myers Squibb: Consultancy; Takeda: Consultancy, Speakers Bureau; Abbvie: Consultancy, Speakers Bureau; Janssen: Consultancy, Speakers Bureau; Teva: Consultancy; TG Therapeutics: Consultancy; Bayer: Speakers Bureau; Sanofi: Speakers Bureau.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4895-4895
Author(s):  
Jonathan How ◽  
Margaret Warner ◽  
Chaim Shustik ◽  
Pierre Laneuville

Abstract Abstract 4895 Background: The treatment of primary central nervous system lymphoma (PCNSL) is characterized by high rates of toxicity and intracranial relapse despite initial response. High-dose methotrexate (HD-MTX), with or without cranial irradiation, has become the standard of care; combining this with other chemotherapeutic agents has improved complete remission rates but at the expense of increased treatment-related adverse events, particularly haematologic and neurologic toxicity. Cytarabine has been used in combination with etoposide (CYVE) as a salvage therapy for patients who have been refractory to or relapsed after HD-MTX-based regimens (Soussain C et al, J. Clin. Oncol. 2001; Soussain C et al, J. Clin. Oncol. 2008). In these studies, cytarabine was given as an infusion at a dose of 50mg/m2/d over 12 hours on days 1–5, and at a dose of 2g/m2/d over 3 hours on days 2–5 with etoposide at 200mg/m2/d on days 2–5. Both responders and non-responders were eligible to proceed to autologous stem cell transplant (autoSCT) using a conditioning regimen of thiotepa 250mg/m2/d on days -9 to -7, busulfan 10mg/kg total dose po or 8mg/kg total dose iv over days -6 to -4, and cyclophosphamide 60 mg/m2/d on days -3 to -2. Median OS was 58.6 months in patients able to undergo autoSCT, but with significant neurotoxicity that may have been related in part to prior therapies. Methods and Results: Given its excellent CNS penetration, CYVE may be an effective regimen in previously untreated patients, with lower risk of neurotoxicity. We have used CYVE as a first-line therapy for patients with PCNSL who are potentially eligible for subsequent autoSCT. Between January 1, 2005 and December 31, 2009, 7 patients with a diagnosis of PCNSL were treated with first-line CYVE using the regimen described above. All were HIV negative, showed no disease outside of the CNS on bone marrow biopsy and staging CT or PET scan, and had biopsy-proven PCNSL with diffuse large B-cell histology. Median age was 55 (42-61) with 4 female and 3 male patients. Median International Extranodal Lymphoma Study Group Prognostic Index score was 2 (0-2), although in actuality may have been higher, as 4 patients were not able to undergo lumbar puncture at diagnosis. ECOG status was < 2 in 3 patients, ≥ 2 in 3 patients, and not documented in 1 case. Patients received a median of 2 cycles of CYVE (1-3), with 2 receiving concurrent rituximab and the 1 patient with intra-ocular lymphoma receiving post-adjuvant orbital radiation. No cranial radiation was administered. Five patients achieved CR and proceeded to autoSCT. Two patients had a PR but later died of progressive disease at day +48 and day +117, respectively. In both cases, however, the patients were suspected of having disease long before they were treated with CYVE. The first had presented 3 years prior to the diagnosis with a solitary brain lesion that resolved with decadron monotherapy before pathologic documentation. The second had had a brain biopsy that failed to show malignant disease 4 months before the PCNSL diagnosis was confirmed and treatment initiated. Median OS and PFS for the group have not yet been reached with mean follow-up of 657 days. Among the 5 patients who achieved CR, all patients remain alive in CR with a mean follow-up of 841 days (320-1262 days), or 28 months. Toxicities included at least 1 episode of febrile neutropenia in all 7 patients, 2 possible but unconfirmed cases of pulmonary aspergillosis, 2 cases of drug-induced hepatitis (with ALT > 3x ULN), 1 case of toxic epidermal necrolysis secondary to imipenem use during an episode of febrile neutropenia, 1 small bowel obstruction requiring surgical intervention, and 1 case of late grade 2 neurotoxicity presenting 3 years after treatment. Conclusions: In this small cohort of patients, first-line CYVE has shown to be highly effective (ORR 100%, CR 71%) with minimal long-term toxicity compared to that which is seen using it as a salvage therapy. When followed by autoSCT, long-term survival with preserved quality of life may be possible. A prospective study would be necessary to confirm these initial results in practice. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 35 (Supplement_3) ◽  
Author(s):  
Roberta Fenoglio ◽  
Martina Cozzi ◽  
Savino Sciascia ◽  
Emanuele De Simone ◽  
Giulio Del Vecchio ◽  
...  

Abstract Background and Aims IgA-vasculitis (IgAV) is a systemic small vessels vasculitis characterized by deposition of underglycosylated IgA1 immune complexes. Renal involvement indicates severity of illness and chronic kidney disease represents the most serious long-term complication of IgAV. Presently, no treatment is specifically recommended in IgAV Glucocorticoids (GC) have been traditionally thought to be effective in tempering systemic symptoms, but did not show long-term benefits either in reducing flares or progression of kidney disease. The effectiveness of conventional immunosuppressants is controversial. Recently Rituximab (RTX) has been proved to be effective in a few case series of adults with IgAV. However, long term results are lacking. Aim of the study: to evaluate the effectiveness of RTX as first line therapy in induction and maintenance of remission of adults with IgAV with biopsy-proven crescentic glomerulonephritis. Method We reviewed the clinical records of patients with adult-onset IgAV treated with RTX at our Center. Patients included 8 males and 4 females, mean age 45 years (range 19-75) with mean follow-up duration of 31 months (range 6-144). Diagnosis was based on the combination of clinical assessment, serological tests and histological analysis according to EULAR criteria. All patients (pts) had a biopsy proven IgAV- severe nephritis. RTX was given for the treatment of relapsing or refractory disease or because of definite contraindications to standard dose CS and/or conventional immunosuppressants. Patients received 4 weekly doses of RTX (375 mg/m2) given alone (8 pts) or in combination with CS (4 pts). Disease activity was evaluated by Birmingham Vasculitis Activity Score version 3 at the onset and at 1, 6 and 12 months and at the end of follow up. Complete remission (CR) was defined as BVAS of 0 Results Eleven pts (91.7%) achieved a clinical response at 6 months. Ten pts had a CR while 1 pt had a partial response and was given an additional dose of RTX after 12 months from induction due to persistent proteinuria (1gr/24 hrs), despite systemic remission. He achieved a CR 6 months later. One patient did not respond to RTX and was switched to MMF. Among the 10 pts with CR, 1 patient needed maintenance doses of RTX every 6 months due to relapse of palpable purpura; 1 relapsed after 15 months and received a new induction course showing a CR again. Significant decrease in 24-hour proteinuria (P = 0.043), BVAS (P = 0.031),and CRP level from RTX initiation through the last follow-up visit was detected. RTX was generally well tolerated. One patient, who had a CR with RTX alone died after 6 months of follow-up for cardiovascular cause. Conclusion This extended experience confirms our initial results supporting the use of RTX in the treatment of IgAV with severe renal involvement. Indeed, RTX proved to be effective and safe for induction and maintenance of long-lasting remission. Present data also suggest that RTX is not only effective for severe and refractory IgAV, but can be also proposed as a first line therapy.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 149-149 ◽  
Author(s):  
Julie E. Chang ◽  
Christopher Peterson ◽  
Lakeesha L. Carmichael ◽  
KyungMann Kim ◽  
David T. Yang ◽  
...  

Abstract Introduction: There remains no clear standard first-line therapy for MCL. VcR-CVAD is a novel, intermediate-intensity chemoimmunotherapy regimen which incorporates bortezomib into modified hyper-CVAD chemotherapy. We hypothesized that the addition of bortezomib would improve the complete response (CR) rate, and maintenance rituximab (MR) would improve the remission duration. The results of this study were previously reported (Chang JE, et al. Br J Haematol 2011), with an observed overall response rate (ORR) of 90% (CR/unconfirmed CR in 77%), and 3-year progression-free survival (PFS) and overall survival (OS) of 63% and 86%, respectively. Long-term follow-up (LTFU) is reported from this multicenter trial. Methods: The study enrolled patients ≥18 years of age with histologically confirmed MCL. Patients were previously untreated, with the exception of 1 cycle of CHOP/CHOP-like chemotherapy. Patients received VcR-CVAD induction chemotherapy every 21 days for 6 cycles: rituximab (R) 375 mg/m2 intravenously (IV) on day 1; bortezomib/Velcade® (Vc) 1.3 mg/m2 IV, days 1 & 4; cyclophosphamide 300 mg/m2 IV every 12 hours, days 1-3 (total of 6 doses); doxorubicin 50 mg/m2 IV continuous infusion days 1-2 (total dose over 48 hours equal to 50 mg/m2); vincristine 1 mg IV day 3; and dexamethasone 40 mg orally days 1-4. Patients received G-CSF support beginning day 5-6 of each induction cycle, and all appropriate supportive care measures were permitted throughout treatment including tumor lysis prophylaxis, transfusion support and antibiotics. Patients achieving at least a partial response to induction therapy received R consolidation (R 375 mg/m2 IV X 4 weekly doses) and MR (R 375 mg/m2 IV every 12 weeks for a total of 5 years; total of 20 doses). Restaging CT scans were performed after cycles 2, 4, and 6 of induction, 12 weeks after consolidation, every 6 months during maintenance, and yearly during LTFU. The primary endpoint was ORR and CR to induction chemotherapy; secondary endpoints were PFS and OS. Results: Thirty patients were enrolled from 7/2005-5/2008. Median age was 61 years (range 48-74), 80% male, all patients had advanced stage disease, and 60% had MIPI score of medium- or high-risk disease. Six patients had blastic morphology. Long-term results are reported after a median follow-up of 7.8 years in surviving patients. Twenty patients are alive, and 15 (50%) are alive in ongoing remission (Figure 1). Estimates of 6-year PFS and OS are 53.1% and 69.8%, respectively (Table 1). The observed PFS and OS differences between patients <age 60 and those ≥age 60 were not statistically significant. The observed PFS and OS differences by MIPI score were not statistically significant, although there was a trend towards worse PFS and OS for high-risk MIPI patients. Five patients have died from confirmed progression of MCL. Two deaths occurred from complications post-allogeneic transplant, and 3 deaths occurred from unrelated causes with MCL in remission. No MCL relapses have been observed beyond 5 years. No late toxicities from VcR-CVAD or from MR have emerged during the LTFU. Conclusions: VcR-CVAD is a moderate intensity chemotherapy regimen that is tolerable for many older and less fit adult patients as first-line therapy of MCL. LTFU of patients receiving VcR-CVAD induction followed by 5 years of MR demonstrates high rates of durable remission that are comparable with more intensive chemotherapy and consolidative autologous stem cell transplant (ASCT). The highly promising activity of the VcR-CVAD regimen was recapitulated in ECOG-ACRIN protocol E1405 (Chang et al, Blood 2014). A randomized phase 3 trial has recently confirmed the beneficial effects of bortezomib incorporation into standard immunochemotherapy (Robak et al, NEJM 2015). VcR-CVAD remains an effective therapy choice for initial treatment of MCL, both in younger and older MCL populations. Disclosures Kahl: Celgene: Consultancy; Seattle Genetics: Consultancy; Infinity: Consultancy; Gilead: Consultancy; Juno: Consultancy; Pharmacyclics: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document