ALBERT

All Library Books, journals and Electronic Records Telegrafenberg

feed icon rss

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
Collection
Years
  • 1
    Publication Date: 2019-11-13
    Description: Introduction: Smoking is a potential risk factor for the development of non-Hodgkin lymphoma (NHL), and prior studies have reported inferior survival in tobacco users with certain subtypes of the disease (Taborelli et al, BMC Cancer, 2017; Ollberding et al, Br J Haematol, 2013). For instance, tobacco smokers with NHL had an inferior overall survival (OS) compared to non-smokers in a series of 471 patients who were managed up front with either chemotherapy (68%), radiation (27%), or observation, and this appeared to be most pronounced in patients with follicular lymphoma and in those with a 20+ pack year smoking history (Geyer et al, Cancer, 2010). The impact of tobacco use on survival specifically in patients with mantle cell lymphoma (MCL) has not been well studied. We conducted a multicenter study in MCL and evaluated the prognostic impact of tobacco use. Methods: We included patients with MCL from 12 sites who were ≥18 years old and for whom smoking status was known at the time of diagnosis. Cases were evaluated for reported smoking status at the time of diagnosis (active smoker, prior smoker, or never smoker) and standard baseline clinical prognostic data were obtained for each patient. Descriptive statistics were generated for these characteristics and were then compared across smoking status using chi-squared tests, Fisher's exact tests, or ANOVA, where appropriate. Overall survival (OS) and progression free survival (PFS) were estimated using the Kaplan-Meier method, and were compared using log-rank tests. Results: Of 946 included patients, 456 (48.2%) reported never using tobacco, 360 (38.7%) reported prior tobacco use, and 130 (13.7%) reported active tobacco use at the time of diagnosis. Median age was 59 in the active smoker group, 65 in prior smokers, and 61 in never smokers (p 〈 0.001). Any major medical comorbidity (defined as the presence of CAD, CHF, diabetes, CKD, ESRD, COPD, DVT, prior malignancy, or cirrhosis) was present in 59 (45.4%) of the active smokers, 143 (39.7%) of the prior smokers, and 140 (30.7%) of the never smokers (p = 0.002). Intensive induction regimens were used in 58.2% of active smokers, 47.2% of prior smokers, and 58.4% of never smokers (p=0.007). There were no significant differences between groups in regards to sex, race, ECOG performance status, Ann Arbor stage, time to first treatment, and use of auto transplant in first remission. Patients with no prior history of tobacco use were less likely to have a high risk MIPI score at diagnosis (26% high risk) compared to prior smokers (39.5%) and active smokers (32.5%, p=0.019). With a median follow up of 3.5 years after diagnosis, there was no significant difference between the 3 groups with regards to PFS or OS (Figure 1). Five-year OS in the never smoker group was 79.8% (95% CI: 74.8%, 83.9%) vs 75.1% (64.5%, 82.9%) in the active smoker group, and 80.6% (74.6%, 85.3%) in the prior smoker group (log rank p = 0.4079). Five- year progression free survival was 50.4% (44.6%, 56.0%) in the never smoker group, 42.5% (32.2%, 52.5%) in the active smoker group, and 50.2% (43.5%, 56.6%) in the prior smoker group (log rank p= 0.3595). Conclusions: Our data suggest that active or prior smoking does not significantly impact OS or PFS in patients with MCL. This study is limited by the fact that amount of current or former tobacco use was not available and it is not known how many current tobacco users ultimately stopped smoking during the course of their treatment. Future studies should incorporate more specific information regarding smoking history including pack-years and time between discontinuation of tobacco use and date of diagnosis. While tobacco use and other modifiable cardiovascular risk factors should be addressed as appropriate for all patients with MCL, current and former tobacco users can still achieve prolonged PFS and OS and may be candidates for intensive treatments after consideration of their other comorbidities and disease-specific risk factors. Disclosures Calzada: Seattle Genetics: Research Funding. Kolla:Amgen: Equity Ownership. Bachanova:Gamida Cell: Research Funding; GT Biopharma: Research Funding; Seattle Genetics: Membership on an entity's Board of Directors or advisory committees; Incyte: Research Funding; Celgene: Research Funding; Novartis: Research Funding; Kite: Membership on an entity's Board of Directors or advisory committees. Gerson:Seattle Genetics: Consultancy; Abbvie: Consultancy; Pharmacyclics: Consultancy. Barta:Janssen: Membership on an entity's Board of Directors or advisory committees; Celgene: Research Funding; Mundipharma: Honoraria; Janssen: Membership on an entity's Board of Directors or advisory committees; Celgene: Research Funding; Takeda: Research Funding; Merck: Research Funding; Mundipharma: Honoraria; Bayer: Consultancy, Research Funding; Seattle Genetics: Honoraria, Research Funding. Danilov:Celgene: Consultancy; Abbvie: Consultancy; TG Therapeutics: Consultancy; Bayer Oncology: Consultancy, Research Funding; Gilead Sciences: Consultancy, Research Funding; Janssen: Consultancy; AstraZeneca: Consultancy, Research Funding; Genentech: Consultancy, Research Funding; Aptose Biosciences: Research Funding; Bristol-Meyers Squibb: Research Funding; MEI: Research Funding; Pharmacyclics: Consultancy; Verastem Oncology: Consultancy, Other: Travel Reimbursement , Research Funding; Curis: Consultancy; Takeda Oncology: Research Funding; Seattle Genetics: Consultancy. Grover:Seattle Genetics: Consultancy. Karmali:Astrazeneca: Speakers Bureau; Takeda, BMS: Other: Research Funding to Institution; Gilead/Kite; Juno/Celgene: Consultancy, Speakers Bureau. Hill:Seattle Genetics: Consultancy, Honoraria; Takeda: Research Funding; Amgen: Research Funding; TG therapeutics: Research Funding; AstraZeneca: Consultancy, Honoraria; Abbvie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celegene: Consultancy, Honoraria, Research Funding; Genentech: Consultancy, Research Funding; Kite: Consultancy, Honoraria; Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Pharmacyclics: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Ghosh:Pharmacyclics: Consultancy, Research Funding, Speakers Bureau; Seattle Genetics: Consultancy, Speakers Bureau; Genentech: Research Funding; Celgene: Consultancy, Research Funding, Speakers Bureau; Forty Seven Inc: Research Funding; Gilead/Kite: Consultancy, Speakers Bureau; Spectrum: Consultancy, Speakers Bureau; AbbVie: Consultancy, Speakers Bureau; T G Therapeutics: Consultancy, Research Funding; Astra Zeneca: Speakers Bureau. Park:BMS: Consultancy, Research Funding; Rafael Pharma: Membership on an entity's Board of Directors or advisory committees; G1 Therapeutics: Consultancy; Teva: Consultancy, Research Funding; Gilead: Speakers Bureau; Seattle Genetics: Research Funding, Speakers Bureau. Epperla:Pharmacyclics: Honoraria; Verastem Oncology: Speakers Bureau. Hamadani:Pharmacyclics: Consultancy; ADC Therapeutics: Consultancy, Research Funding; Merck: Research Funding; Celgene: Consultancy; Janssen: Consultancy; Medimmune: Consultancy, Research Funding; Sanofi Genzyme: Research Funding, Speakers Bureau; Otsuka: Research Funding; Takeda: Research Funding. Kahl:TG Therapeutics: Consultancy; BeiGene: Consultancy; Seattle Genetics: Consultancy; ADC Therapeutics: Consultancy, Research Funding. Martin:Janssen: Consultancy; Sandoz: Consultancy; I-MAB: Consultancy; Teneobio: Consultancy; Celgene: Consultancy; Karyopharm: Consultancy. Flowers:Karyopharm: Consultancy; Denovo Biopharma: Consultancy; Burroughs Wellcome Fund: Research Funding; AbbVie: Consultancy, Research Funding; Gilead: Consultancy, Research Funding; Spectrum: Consultancy; AstraZeneca: Consultancy; Pharmacyclics/Janssen: Consultancy, Research Funding; Bayer: Consultancy; Acerta: Research Funding; Genentech, Inc./F. Hoffmann-La Roche Ltd: Consultancy, Research Funding; Optimum Rx: Consultancy; Millenium/Takeda: Research Funding; Eastern Cooperative Oncology Group: Research Funding; National Cancer Institute: Research Funding; V Foundation: Research Funding; BeiGene: Consultancy, Research Funding; Celgene: Consultancy, Research Funding; TG Therapeutics: Research Funding. Cohen:Genentech, Inc.: Consultancy, Research Funding; Janssen Pharmaceuticals: Consultancy; Takeda Pharmaceuticals North America, Inc.: Research Funding; Gilead/Kite: Consultancy; LAM Therapeutics: Research Funding; UNUM: Research Funding; Hutchison: Research Funding; Astra Zeneca: Research Funding; Lymphoma Research Foundation: Research Funding; ASH: Research Funding; Seattle Genetics, Inc.: Consultancy, Research Funding; Bristol-Meyers Squibb Company: Research Funding.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 2
    Publication Date: 2018-11-29
    Description: 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.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 3
    Publication Date: 2019-11-13
    Description: Introduction: Time to treatment (TTT) is an important prognostic factor in patients with newly diagnosed diffuse large B-cell lymphoma (Maurer et al JCO 2018) where patients who initiate therapy quickly after diagnosis have an inferior event free survival compared to those who do not require such immediate treatment initiation. More recently, TTT has shown to be associated with adverse clinical factors and inferior outcomes in mantle cell lymphoma (MCL; Maurer, et al ASH, 2018). However, there is a paucity of data on the impact of TTT on overall survival (OS). We sought to validate these findings and to evaluate the impact of TTT on survival outcomes in newly diagnosed patients with MCL. Methods: We included patients from 12 medical centers in the United States with MCL diagnosed between January 1, 2000, and January 1, 2018, who had information on the TTT and initiated treatment within 60 days of diagnosis (to exclude patients whose treatment was purposefully deferred). TTT was defined as the time in days from first lymphoma diagnosis to initiation of therapy. Patients who received treatment within 14 days were categorized into short TTT group. We compared differences between the two groups (TTT 14 days and ≤ 60 days, longer TTT group) using chi-squared test, Fisher's exact tests, or ANOVA tests as appropriate. OS was defined as the time from diagnosis to death or last follow-up. Patients not experiencing an event were censored at their last known follow-up. OS was determined using the Kaplan-Meier method, and univariable and multivariable models were developed to identify predictors of OS. Results: Of 1,168 patients with newly diagnosed MCL, seven hundred fifty-five patients met the inclusion criteria and were included in this analysis, including 205 (27%) with short TTT and 550 (73%) with longer TTT. Median time to treatment was 7 days (range, 0-14) for the short TTT group vs 31 days for the longer TTT group (range, 15-60 days). The median age for the entire cohort was 63 years, 75% of patients were male, and 93% of patients had ECOG 0-1. The proportion of patients with stage 4 disease (93 vs 86%, p=0.015), elevated LDH (58 vs 39%, p3 cytogenetic abnormalities (29 vs 14%, p=0.005), B symptoms (47 vs 29%, p
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 4
    Publication Date: 2018-11-29
    Description: Introduction Mantle cell lymphoma (MCL) frequently impacts elderly patients (median age of diagnosis is 68), and management of these patients can be difficult due to the presence of comorbidities in this group. Presence of comorbidities is associated with inferior outcomes in many hematologic malignancies but there has not been a systematic assessment of comorbidities in MCL. We utilized the Charlson Comorbidity Index (CCI) to describe the presence of comorbidities in MCL patients and to explore their impact on patient outcomes. Methods We included patients with MCL evaluated at Emory between January 1, 2000 and December 31, 2016. Patients with limited or inadequate follow-up or for whom data to determine the CCI were inadequate were excluded. We determined the CCI at the time of initiation of treatment for all patients with MCL and excluded "lymphoma" from this calculation. Patients were categorized into severity classes based on their summed scores. The median score of 1 was used to delineate the "low" and "high" CCI groups. For each group (low and high CCI), we evaluated baseline demographic, clinical and treatment characteristics and identified specific toxicity-related outcomes including: failure to complete induction chemotherapy, unplanned hospitalizations, or treatment related toxicities. Progression-free (PFS) and overall survival (OS) were estimated using the Kaplan-Meier method, and we assessed for the impact of CCI score on PFS and OS using Cox proportional hazards models. Results Of 178 patients with MCL, 129 were male (73.3%) and median age was 61 (32-86). One hundred forty patients (86.4%) had stage IV disease. Among the 99 reported values, 31 patients (31.3%) had an elevated LDH, and ECOG PS was 0 in 47 patients, 1 in 66 patients, and 2-4 in 8 patients. Induction therapy was R-HyperCVAD (n=79; 45.4%), R-CHOP (n=49; 28.2%), BR (n=13; 7.5%) RCHOP/RDHAP (n=3; 1.7%) and Other (N=29; 16.7%). The CCI scores for the whole cohort were 0 in 78 pts (43.8%), 1 in 57 pts (32%), 2 in 24 pts (13.5%) and 3+ in 19 pts (10.7%; See Figure 1). One hundred thirty-five patients (75.8%) patients had a low CCI score (0-1) and 43 patients (24.2%) had a high CCI score (〉1). Compared to patients with a low CCI, those with high CCI were older (median 67 years vs 59 years, p 0.3). Conclusion CCI score was not shown to be a reliable predictor of unplanned hospitalizations, premature cessation of chemotherapy, or OS in our cohort, although many of our patients had very limited comorbidities. These results suggest that patients with limited comorbid conditions can likely be successfully managed with appropriate supportive care and should be considered for the most effective regimens. Future studies with a larger patient population with increased number of comorbidities may improve our ability to detect the impact of CCI on these outcomes and explore the relative contribution of specific comorbidities in MCL. Disclosures Calzada: Seattle Genetics: Research Funding. Flowers:BeiGene: Research Funding; Burroughs Wellcome Fund: Research Funding; Abbvie: Consultancy, Research Funding; Abbvie: Research Funding; Pharmacyclics/ Janssen: Consultancy; Millennium/Takeda: Research Funding; Gilead: Consultancy; Genentech/Roche: Consultancy; Janssen Pharmaceutical: Research Funding; Genentech/Roche: Research Funding; OptumRx: Consultancy; Acerta: Research Funding; Gilead: Research Funding; Denovo Biopharma: Consultancy; Celgene: Research Funding; Bayer: Consultancy; Spectrum: Consultancy; Karyopharm: Consultancy; National Cancer Institute: Research Funding; TG Therapeutics: Research Funding; Eastern Cooperative Oncology Group: Research Funding; Pharmacyclics: Research Funding; V Foundation: Research Funding. Cohen:AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Millennium: Consultancy, Membership on an entity's Board of Directors or advisory committees; Infinity Pharmaceuticals: 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; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Research Funding; Infinity Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Research Funding; AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Research Funding; Bristol-Myers Squibb: Research Funding; Millennium: 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; Novartis: 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; Pharmacyclics: Consultancy, Membership on an entity's Board of Directors or advisory committees; Takeda: Research Funding; Takeda: Research Funding; BioInvent: Consultancy; BioInvent: Consultancy; Seattle Genetics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Seattle Genetics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 5
    Publication Date: 2018-11-29
    Description: Introduction: While most patients with mantle cell lymphoma (MCL) receive therapy shortly after diagnosis, a subset of patients with indolent-behaving disease can safely defer treatment (Calzada et al, 2018). In this subgroup, a lack of evidence to support the decision-making behind the choice of therapy has led to a wide diversity in treatments. We evaluated the importance of treatment intensity in patients with MCL who defer initial therapy. Methods: We included patients ≥ 18 years old with MCL from 11 academic centers and defined the deferred subgroup as patients who started therapy ≥ 90 days after diagnosis. Patients who received high dose cytarabine as part of their induction or autologous stem cell transplantation (ASCT) in first remission were considered to have received intensive therapy while all other approaches were non-intensive. We identified differences between the baseline characteristics of the two groups using Fisher's exact tests, chi-squared tests, and t-tests as appropriate. We calculated progression-free (PFS) and overall survival (OS) from the date of diagnosis using the Kaplan-Meier method and compared the two groups using the log-rank test. Univariate and multivariate Cox proportional hazards models were performed for PFS and OS. Results: Of 968 identified patients with MCL, 233 did not initiate therapy within 90 days of diagnosis and were considered deferred. Deferred patients had a lower Ann Arbor stage (p
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 6
    Publication Date: 2019-11-13
    Description: Background: Early progression of disease (POD) in patients (pts) with mantle cell lymphoma (MCL) following intensive frontline treatment has been associated with inferior survival (Dietrich et al Ann Oncol 2014 and Visco et al Br J Haematol 2019), but the optimal time point to define early POD and the predictive significance of early POD following less intensive frontline treatment is not well established. We compare outcomes after all frontline treatments in a large MCL cohort categorized by time to progression and describe outcomes by class of second line treatment for pts with primary refractory disease. Methods: Clinical and outcome data for MCL pts treated between 2000 and 2017 were collected from 12 US centers. Overall survival (OS), defined from time of 1st progression, and secondary progression free survival (PFS2), defined from 1st progression to 2nd progression or death, were estimated by Kaplan-Meier and compared by log-rank test. Univariable and multivariable analyses were performed using Cox proportional hazards models for OS. 95% confidence intervals were calculated for all estimates and displayed in square brackets. We defined intensive treatment as high dose cytarabine in frontline therapy and/or autologous stem cell transplant in 1st remission. Pts were categorized into three groups: (a) refractory disease to frontline therapy or POD within 6 months of frontline therapy was termed primary refractory (PRF); (b) POD between 6 to 24 months of therapy was termed POD24 and (c) POD beyond 24 months was termed POD〉24. Salvage therapy was categorized as chemoimmunotherapy (CIT) for pts treated with CIT alone, BTK inhibitor (BTKi) for pts treated with BTKi single agent or in combination, and lenalidomide / bortezomib for pts treated with one or both agent +/- anti-CD20 therapy. Results: Of 1,168 pts with MCL, 457 pts had relapsed and were included in this analysis. The median age was 62, 77% were male, and ECOG PS was 0/1 in 94%. Median follow-up was 2.6 years (yr) after first progression. The most common induction regimens were R-HyperCVAD (26%), R-CHOP (24%), bendamustine and rituximab (19%), and R-M-CHOP (10%). Frontline treatment was intensive in 54%. Sixty five pts (14%) were PRF, 153 (34%) had POD24, and 239 (53%) had POD〉24. Additional baseline characteristics and comparison between groups are summarized in Table 1. The median OS was 1.3 yr [0.9-2.4] for PRF pts, 3 yrs [2-6.8] for POD 24, and 8 yrs [6.2-not reached (NR)] for POD〉24 (p24 pts (p
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 7
    Publication Date: 2019-11-13
    Description: INTRODUCTION The optimal frontline treatment for mantle cell lymphoma (MCL) is not clearly defined. Bendamustine + rituximab (BR) is commonly used as initial therapy. The role of maintenance rituximab (MR) after BR is not agreed upon due to limited data supporting this practice, whereas MR improves overall survival (OS) after autologous stem cell transplant (ASCT) and after R-CHOP for elderly patients who do not receive ASCT. Preliminary results from a subgroup analysis of the randomized phase 3 MAINTAIN study revealed neither a progression-free survival (PFS) nor OS benefit for MR as compared to observation for MCL pts (Rummel, ASCO 2016). In follicular lymphoma patients, however, there does appear to be a PFS benefit to rituximab maintenance following BR. Given these disparate results, we sought additional data to evaluate the role of rituximab maintenance following BR in MCL METHODS MCL pts treated at 12 U.S. medical centers with frontline BR who achieved a complete response (CR) or partial response (PR) and who did not receive consolidative ASCT from 2011 - 2017 were included. Use of MR was based on individual physician/patient preferences. Baseline pt characteristics were compared using chi-squared test, Fisher's exact tests, or ANOVA. Descriptive statistics, comparisons, and OS using the Kaplan-Meier method were stratified by response status as determined by the treating site (complete response (CR) only, partial response (PR) only, and CR/PR). RESULTS Among 135 pts responding to frontline BR who did not complete subsequent ASCT, 80% achieved complete remission (CR) and 20% had a partial remission (PR). Median age was 70 (range 45 - 93) years and 66% were male. Baseline MIPI score was low (13%), intermediate (38%), or high (49%) among patients with available data (n = 92) and did not differ between treatment cohorts. Among responding patients, 78 (58%) received MR and 57 (40%) were observed. With a median follow up of 3.1 years, median OS was not reached for pts responding to BR (with CR or PR) who received MR vs. 6 years for those who received no maintenance (Figure Panel A, P = 0.0013). Use of MR vs. observation was associated with a significant improvement in OS for pts in PR at the end of induction therapy (Figure Panel B, median not reached vs. 1.7 years, P = 0.006), but there was no statistically significant OS difference for pts in CR (Figure Panel C, median not reached vs. 9.6 years, P = 0.2575). In multivariable analysis, MIPI score
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 8
    Publication Date: 2018-11-29
    Description: Background High dose chemotherapy and autologous stem cell transplant (ASCT) results in cure for up to 50% of patients with Hodgkin (HL) and aggressive non-Hodgkin lymphoma (NHL) that relapse after initial treatment with chemo-immunotherapy. Despite improvements in supportive care and patient selection, death after ASCT remains a concern due to progressive disease, infection, and other treatment- and non-treatment-related causes. We evaluated causes and predictors of death in patients undergoing ASCT for HL and NHL. Methods We conducted a single-institution, retrospective study of all patients with HL and NHL including diffuse large B-cell lymphoma, high grade B-cell lymphoma NOS, follicular lymphoma, Burkitt lymphoma, marginal zone lymphoma, and T-Cell lymphoma who underwent ASCT at Emory University between January 1, 2006 and June 1, 2017. We evaluated each patient with regards to pre-transplant disease and other baseline characteristics, treatment response after ASCT, occurrence of relapse post-ASCT, post-ASCT death and cause of death. Among patients who died, we compared baseline characteristics of interest between groups based on the documented cause of death using Fisher's Exact tests and t-tests. The association between cause of death and time to death from ASCT was evaluated by the Kruskal-Wallis test. Box plots were used to describe time to death post-transplant. Results Of 642 patients completing ASCT, 192 died during the period of observation after a median follow-up of 24 months. Among patients who died, the leading causes of death were relapsed disease (n=136, 71%), infection (n=18, 9%), organ dysfunction (n=16, 8%), secondary malignancy (n=8, 4%), and other/unspecified (n-14, 7%). When the group was divided into disease-related (n=136) vs non-disease-related death (n=56), none of the pre-specified variables of interest including age, gender, disease status at transplant, stage, induction or salvage therapy, conditioning regimen, or KPS were significantly associated with disease- vs non-disease-related death. Of the 18 deaths due to infection 7 were due to bacterial sepsis (including 5 prior to day +30 post ASCT), 3 were due to fungal infections, 3 were due to pneumonia, 1 death each was due to hepatitis C, nocardia, pneumocystis jirovecii, cytomegalovirus, and C. Difficile. The median time to death for patients experiencing infection or organ dysfunction was 0.5 years each compared to 1 year for patients dying of disease relapse and 4 years for patients experiencing a secondary malignancy (p
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 9
    Publication Date: 2019-11-13
    Description: Introduction: Mantle cell lymphoma (MCL) is a rare form of lymphoma with no current standard of care. As a result, clinical trials are critical to improving our understanding of the disease and its management. However, clinical trial participation is often limited due to lack of access to studies, restrictive eligibility criteria, and decisions by treating physicians or patients not to participate. We evaluated the rate of enrollment and outcomes associated with clinical trial participation of patients with MCL at 12 US Medical Centers. Methods: We included patients from 12 medical centers in the United States with MCL diagnosed between January 1, 2000, and January 1, 2018, who were ≥ 18 years old, received any induction therapy, and for whom it was known whether initial treatment was on a clinical trial (CT) or not. We compared differences between the two groups (CT vs not) using Fisher's Exact and Chi-square tests as appropriate. We defined overall survival (OS) as time from diagnosis to death. Patients not experiencing an event were censored at their last known follow-up. OS was determined using the Kaplan-Meier method, and univariate and multivariable models were developed to identify predictors of OS. Results: Eight hundred twelve patients were included in this analysis, including 164 (20%) patients who participated in a clinical trial during initial therapy. Rate of participation ranged from 4% to 36% across contributing sites. Median age for the entire cohort was 62 years, 572 (74%) of patients were male, and 95% of patients had ECOG 0-1. MCL International Prognostic Index (MIPI) risk score was high in 163 patients (31%), and 84% of patients had stage IV disease. Ki67 was 〉30% in 169/225 (43%) patients with available data, and 19% of patients (65/271) had a complex karyotype with 〉 3 chromosomal abnormalities. Four hundred sixteen (53%) of patients received an intensive induction regimen (defined as initial treatment with a high-dose cytarabine-containing regimen and/or receipt of autologous hematopoietic cell transplantation as consolidation), including 333 patients (43%) who underwent transplant consolidation. Patients with an increased lactate dehydrogenase (LDH) level (p 3 cytogenetic abnormalities (p=0.015) were less likely to participate in clinical trials, but there was no significant difference in rates of participation based on MIPI (p=0.49) or ECOG performance status (p=0.22; Table). Patients treated on study were less likely to receive an intensive regimen compared to those treated off study (p=0.002). Median time to treatment from diagnosis was 35 days for patients enrolled on trial and 31 days for patients not enrolled on trial (p=0.83). With a median follow-up of 3.8 years, the median OS was 13.6 years (95% CI: 11.5-21.1) for patients not treated on a trial and not reached (95% CI: 9.9 - Not Reached) for patients treated on trial (Figure; p=0.036). In a multivariable model including clinical trial participation, MIPI, time to initial treatment, and receipt of an intensive induction regimen, only clinical trial participation (HR 0.54, 95%CI: 0.31-0.93; p=0.028) and high risk MIPI score (HR 4.24, 95% CI: 2.37-7.56; p
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 10
    Publication Date: 2020-11-05
    Description: Introduction Sézary syndrome (SS) is an aggressive, leukemic subtype of cutaneous T-cell lymphoma (CTCL) with a median survival of 3-5 years. Approved therapies include skin-directed therapy, radiation, and systemic therapies such as chemotherapy, histone deacetylase (HDAC) inhibitors, interferon, extracorporeal photopheresis (ECP), and oral retinoids. There is no consensus first-line therapy for SS and there is limited data regarding prognostic biomarkers. We assessed treatment patterns, outcomes, and racial differences at our institution. Methods We performed a retrospective review of 62 patients at Winship Cancer Institute of Emory University from 1990-2020 with a confirmed diagnosis of SS. Clinical data collected from the electronic medical record included demographics, baseline laboratory values, disease characteristics, and therapy. Clinical outcomes were measured by overall survival (OS) and time to next treatment (TTNT). OS was measured from time of diagnosis to date of death or last follow-up. TTNT was defined as the number of months from the start of the first line of therapy until the initiation of the subsequent therapeutic regimen. Descriptive analysis was performed for each variable and a comparison between African American (AA) and white patients was performed using ANOVA for numerical covariates and chi-square test or Fisher's exact test for categorical covariates. Kaplan-Meier curves for OS and TTNT were generated for the whole cohort. A Kaplan-Meier curve was also generated to compare time from diagnosis to initiation of first systemic therapy stratified by race along with the log-rank p-value. The univariate association of baseline variables with OS and TTNT was assessed by Cox proportional hazards models and the multivariable analyses (MVA) were performed on variables that had p-value less than 0.05 on univariate analyses. Results Males made up (58.1%) of our patients and the median age at diagnosis was 65.9 years. Nearly one-half (45.2%) of patients were AA. The median Sézary count at diagnosis was 1320 cells/uL. The median time from diagnosis to first systemic therapy was 2.4 months and the median number of systemic therapies was 3.0. Information regarding systemic treatments received after diagnosis is presented inTable 1. The most common first-line systemic therapies were oral retinoids (43.5%), ECP (32.3%), and interferon (30.6%). HDAC inhibitors and total skin electron beam (TSEB) radiation were common treatments beyond first line (46.8% received HDAC inhibitors, 38.7% received TSEB), but were rarely used in the first-line setting. The median OS and TTNT were 3.1 years and 6.3 months, respectively(Figure 1). In MVA, elevated WBC and LDH were significantly associated with shorter OS (WBC HR: 1.05, 95% CI: 1.01-1.08, p=0.01; LDH HR: 1.003, 95% CI: 1.001-1.005, p=0.011) and shorter TTNT (WBC HR: 1.04, 95% CI: 1.002-1.08, p=0.041; LDH HR: 1.002, 95% CI: 1.001-1.004, p=0.048). In analysis by race, AA patients had a higher proportion of females compared to non-hispanic white patients (53.6% vs 28.1%, p=0.045). AA patients also had lower median hemoglobin at diagnosis (12.6 vs 14.3, p=0.036), higher median LDH at diagnosis (360 vs 232, p=0.002), and longer median time from diagnosis to first systemic therapy compared to non-hispanic white patients (3.17 months vs 2.14 months, p=0.039,Figure 2). Conclusions SS is an aggressive subtype of CTCL with no consensus first-line therapy and limited data on prognostic biomarkers. In our cohort, oral retinoids, ECP, and interferon were the most commonly utilized treatments in the first-line setting. Elevated WBC and LDH were significantly associated with both OS and TTNT which suggests that these may have value as prognostic biomarkers in SS. AA patients may have delayed time from diagnosis to starting systemic therapy and higher LDH at diagnosis. This data is hypothesis-generating and should be validated in larger, prospective studies. Disclosures Allen: Bayer:Consultancy, Other;Imbrium:Consultancy, Other;Research to Practice:Speakers Bureau;Clinical Care Options:Speakers Bureau;Curio Sciences:Honoraria.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...