scholarly journals Improvements in Clinical Outcomes of Advanced Stage Classical Hodgkin Lymphoma in the United States from 2000-2014: Analysis of Surveillance Epidemiology and End Results Database

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2939-2939
Author(s):  
Guru Subramanian Guru Murthy ◽  
Aniko Szabo ◽  
Mehdi Hamadani ◽  
Timothy S. Fenske ◽  
Nirav N Shah

Abstract Background: Advanced Stage Hodgkin lymphoma (HL) is a curable malignancy with combination chemotherapy. While most patients are cured with frontline therapy, for those with refractory disease or early progression, historically, the outcomes have been poor. Novel therapies, PET/CT adapted treatment approaches, and improvement in transplantation have changed the management of both frontline and relapsed HL. However, it remains unknown if these developments have improved the clinical outcomes at population level over time. Methods: Using Surveillance Epidemiology and End Results database, we identified patients aged ≥ 18 years with advanced stage (Stage III or IV) pathologically confirmed classical HL as the first primary malignancy,diagnosed between the years 2000-2014, treated with chemotherapy and actively followed. Patients were stratified by date of diagnosis into 3 groups - 2000-2004, 2005-2009, 2010-2014 to assess the trends in overall survival (OS) over time. Race/ethnicity was stratified into non-hispanic whites and minorities (Non-hispanic blacks, Hispanics, other non-hispanic races). Kaplan-Meier method and log rank test were used to analyze the OS among subgroups. Cox proportional hazard regression method was used to determine the influence of period and demographic factors on OS.Cumulative incidence of death from cardiac cause was estimated using the Nelson-Aalen estimates. Statistical analyses were carried out with significant two sided p< 0.05. Results: A total of 9042 patients with a median age of 41 years were included. There were more males (60.1%) and non-Hispanic whites (64.2%) and most patients had nodal disease (98%) (Table 1).The use of frontline radiation therapy decreased in each 5-year time period (21.3% 2000-2004 vs 15.5% 2005-2009 vs 10.7% 2010-2014, p<0.001). In terms of survival, when stratified by the period of diagnosis, the 3-year OS was significantly higher for patients diagnosed between the year 2010-2014 (81.8%) and 2005-2009 (80.6%) than those diagnosed from 2000-2004 (78.5%, p=0.0008 and 0.02 respectively) (Figure 1). Additionally, age was a significant predictor for OS with a decreasing 3-year OS with increasing age (age < 40 - 91.1%, age 41-60 -81.5%, age >60 -54%, p< 0.001, Figure 2).While outcomes were poorest in the age>60 cohort, similar improvements were seen in OS over the three time periods among this patient population (48.6%- 2000-2004 vs 54.3% 2005-2009 vs 56.8% 2010-2014, p=0.005). On multivariate analysis, diagnosis in the earlier period was associated with higher mortality (2000-2004-HR 1.36, 95% CI 1.21-1.53, p< 0.001; 2005-2009 -HR 1.14, 95% CI 1.01-1.28, p=0.02, both compared to reference group 2010-2014). Similarly, minority races (HR 1.36, 95%CI 1.23-1.49, p<0.001) had a higher mortality risk as compared to non-Hispanic whites. Females (HR 0.82, 95%CI 0.75-0.90 p<0.001) and married status (HR 0.80, 95%CI 0.72-0.87, p< 0.001) were associated with significantly lower mortality. While radiation use decreased over time, the cumulative incidence of cardiac related cause of death did not vary significantly among the three-time periods (1.2% vs 1.1% vs 1.1% respectively at 48 months, p=0.85). Conclusions: Survival of patients with advanced stage HL has continued to improve over time suggesting the clinical impact of evolving treatment approaches. Interestingly this improvement has occurred despite the decreasing utilization of radiation therapy over time. This is suggestive of better end of treatment assessment with PET/CT eliminating the need for end of treatment radiation, improved second line therapies that extend survival, or potentially reduction in treatment related toxicities.Despite these encouraging results, the 3-year OS in the contemporary period remains inadequate at 81.8%. Furthermore, significant differences in survival continue to exist among non-modifiable factors such as gender, age, race, and marital status, highlighting the need for continued research to address these discrepancies. Results of this study provide a new baseline to test novel frontline combination regimens. Disclosures Hamadani: Takeda: Research Funding; Celgene Corporation: Consultancy; Cellerant: Consultancy; Ostuka: Research Funding; Janssen: Consultancy; MedImmune: Consultancy, Research Funding; Merck: Research Funding; ADC Therapeutics: Research Funding; Sanofi Genzyme: Research Funding, Speakers Bureau. Shah:Miltenyi: Other: Travel funding, Research Funding; Exelexis: Equity Ownership; Oncosec: Equity Ownership; Geron: Equity Ownership; Juno Pharmaceuticals: Honoraria; Lentigen Technology: Research Funding.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2832-2832 ◽  
Author(s):  
Herve Ghesquieres ◽  
Olivier Casasnovas ◽  
Emmanuelle Nicolas-Virelizier ◽  
Damaj Gandhi Laurent ◽  
Vincent Delwail ◽  
...  

Introduction: Older patients with an age above 60 years with classical Hodgkin lymphoma (cHL) represent a proportion of 20% to 30% of all cHL. Older cHL patients are characterized by unfavorable prognostic factors with an aggressive disease, a poor tolerance to chemotherapy resulting to a significant reduced survival as compared to younger patients. In relapsed and refractory HL, prospective and retrospective studies showed that bendamustine in monotherapy provided interesting efficacy with 30% of complete response with an acceptable toxicity profile. We developed the PVAB (Prednisone, Vinblastine, Doxorubicin and Bendamustine) regimen in first line therapy to improve prognosis of older HL with advanced stage. Methods: In this prospective phase II, we recruited newly diagnosed classical HL patients age of 61 years or older with an advanced stage (Ann Arbor stage III, IV, IIB with risk factors). Inclusion criteria were: baseline 18-FDG PET scan performed before any treatment with at least one hypermetabolic lesion; ECOG performance status 0-2; adequate cardio-pulmonary function with LVEF ≥ 50%; adequate renal function with creatinine clearance ≥ 40 mL/min; HIV negative; For patients aged 70 years old and more, a Mini Nutritional Assessment (MNA) ≥ 17. Treatment consisted of 6 cycles of prednisone (40mg/m2 D1-5), vinblastine (6mg/m2, D1), doxorubicin (40mg/m2, D1) and bendamustine (120mg/m2, D1) every 21 days. A first evaluation was performed after 4 cycles by CT scan and a final evaluation by PET scan after 6 cycles. No radiotherapy was applied in this protocol. The primary endpoint was the complete metabolic response (CMR) rate after 6 cycles of study treatment or at premature treatment discontinuation according to Lugano Classification. The main analysis for the CMR rate was based on a Simon's phase II design. We selected P0 and P1 to be 70% and 85%, respectively for CMR rate. A total of 79 patients provided nominal power of 80% at the nominal one-sided 5% significance level. Using a drop-out rate of 10%, 90 patients should be included in this trial. Results: Between July 2015 and July 2018, 89 patients who signed the consent form and received at least one PVAB cycle corresponding to intention to treat (ITT) group were included in 34 LYSA centers. Among them, four patients did not respected major inclusion criteria (one patient had a nodular lymphocyte predominant subtype after histological review and three patients ≥ 70 years had no MNA evaluation at inclusion) corresponding to the modified ITT group (N=85). The median age of the 89 patients was 68 years (range, 61-88) with 35 patients ≥70 years old (39%) and 58 male (65%). According to the central review, the main histological subtype was nodular sclerosis cHL (66%). Ann Arbor stages were as follows: II (n=3, 3%), III (n=30, 34%), IV (n=56, 63%). B symptoms were present in 57% of patients and 70 patients (80%) had IPS≥3. 78 patients (88%) completed the 6 cycles of PVAB. In ITT, the CMR rate corresponding to the primary endpoint of the study was 77.5% (95%CI, 67-86) with 69 patients in CMR; 8 patients were in partial metabolic response; 2 and 5 patients had a stable and progressive disease, respectively and 5 were not evaluable. In the modified ITT group, the CMR rate at this end of treatment was 77.6% (95%CI, 67-86). With a median follow-up of 23 months (0.5-40.3), 25 patients relapsed or progressed (28%). The 2-year progression survival rate (PFS) rate was 61.3% (95%CI, 49-72). For the 69 patients achieving CMR, the 2-year disease free survival (DFS) rate was 73.3% (95%CI, 61-82). At the date of analysis (December 2018), 17 patients (19%) died: 7 cHL, 4 treatment toxicity, 3 second cancers, 3 other causes. The 2-year overall survival rate was 84.1% (95%CI, 73-91). For toxicity, 4 patients presented toxic death during treatment: one cardiogenic shock (71y, >cycle 1) one septic shock (70y, >cycle 1), one brain hematoma with grade 4 thrombocytopenia (76y, >cycle 1), one fungal infection (86y, >cycle 4). At least one serious adverse events (SAE) were presented by 28 patients (31.5%) mainly infections (13 patients, 15%), blood (11 patients, 12%) and cardiac disorders (4 patients, 4.5%). Conclusions: Six cycles of PVAB regimen provided high CMR (77.5%) with acceptable toxicity in older cHL patients with advanced stage. Patients with CMR at the end of treatment had a particular favorable outcome but long term follow-up is needed for a better evaluation of survival endpoints. Disclosures Morschhauser: Roche/Genentech: Consultancy; Celgene: Honoraria; Servier: Consultancy; Gilead: Consultancy; Janssen: Honoraria; BMS: Honoraria. André:Celgene: Other: Travel grants, Research Funding; Chugai: Research Funding; Takeda: Membership on an entity's Board of Directors or advisory committees; Bristol-Myers-Squibb: Membership on an entity's Board of Directors or advisory committees; Karyopharm: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees, Other: Travel grants; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Seattle Genetics: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees; Roche: Other: Travel grants, Research Funding; Amgen: Other: Travel grants, Research Funding; Johnson & Johnson: Research Funding; Takeda Millenium: Research Funding. Quittet:Novartis: Honoraria, Speakers Bureau. Brice:Takeda France: Consultancy, Honoraria; Millennium Takeda: Research Funding; BMS: Honoraria. OffLabel Disclosure: Bendamustine is off-label drug use in Hodgkin lymphoma


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1284-1284
Author(s):  
Kailash Mosalpuria ◽  
Fausto R Loberiza ◽  
R. Gregory Bociek ◽  
Matthew A. Lunning ◽  
Apar Kishor Ganti ◽  
...  

BACKGROUND: With improvements in the treatment of Hodgkin lymphoma (HL) over time, we hypothesized a reduction in disease progression, and an increase in progression-free (PFS) and overall survival (OS) over the past 3 decades. METHODS: This is a retrospective study that included previously untreated HL patients reported to the Nebraska Lymphoma Study Group between 1983 and 2012. To analyze trends in outcomes, we divided the cohort into 3 time periods of 10 years each (1983-1992, 1993-2002, 2003-2012) to represent early (E), middle (M) and late (L) time periods. Patient, disease, and treatment-related factors as well as clinical outcomes (cumulative incidence of progression, PFS and OS) were compared according to time period. Multivariate analyses (MVA) were performed using Cox regression to adjust for significant covariates. RESULTS: In this retrospective review, 528 patients were identified during the study period: E (n=120), M (n=208), L (n=200). Patients in the more recent cohort (L) were more likely to be younger (p=0.008), have better performance score (p<0.001), have more sites of nodal involvement (p=0.05) and were more likely to receive ABVD (doxorubicin, bleomycin, vinblastine, dacarbazine) or Stanford V (mechlorethamine, doxorubicin, vinblastine, vincristine, bleomycin, etoposide, prednisone) as initial therapy (p<0.0001). In univariate analysis, there was a decrease in the 5-year cumulative incidence rate of disease progression over time (E=27% vs M=22% vs L=21%), and an increase in the 5-year probability of PFS (E=58% vs M=66% vs L=74%, p=0.004) and 5-year OS (E=67% vs M=80% vs L=85%, p=0.001). In MVA, improvement in the cumulative incidence of disease progression, PFS and OS over time was mainly driven by prognostic patient-related (age, sex) and treatment-related factors (type of chemotherapy regimen). Independent predictors of shorter PFS included age ≥45 y and male gender (Hazard Ratio [HR] 2.00 [95% CI 1.52-2.62, p<0.001] and 1.43 [1.09-1.87, p<0.01], respectively), while patients who received ABVD and Stanford V regimens as initial therapy experienced improved PFS (HR 0.39 [0.23-0.65, p<0.001] and 0.42 [0.28-0.64, p<0.001], respectively versus MOPP-like regimens). For OS, patients who were ≥45 y and males were more at risk of death (HR 4.34 [3.08-6.08, p<0.001] and 1.73 [1.26-2.37, p<0.001], respectively) while patients who received ABVD and Stanford V regimens as initial therapy were less likely to die (HR 0.36 [0.19-0.69, p <0.01] and 0.39 [0.23-0.65, p<0.001], respectively compared to MOPP-like regimens). Similarly, in MVA, a decrease in risk of progression was noted for patients who received ABVD and Stanford V regimens (HR 0.36 [0.19-0.69, p<0.01] and 0.39 [0.23-0.65, p<0.001], respectively) compared to MOPP-like regimens, while increased for patients who were ≥45 y and male (HR 4.33 [3.08-6.08, p<0.001] and 1.73 [1.26-3.37, p<0.001], respectively). CONCLUSIONS: Changes in upfront treatment regimens for HL over the last 3 decades have resulted in significant improvement in PFS and OS, and modest improvement in the risk for progression. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2704-2704 ◽  
Author(s):  
Ted Wun ◽  
Marilyn J. Telen ◽  
Lakshmanan Krishnamurti ◽  
Timothy L. McCavit ◽  
Laura M DeCastro ◽  
...  

Abstract Introduction: The pan-selectin antagonist rivipansel (GMI-1070) reduced intravascular arrest of red/white blood cell aggregates and improved blood flow and survival in a mouse model of sickle cell disease vaso-occlusive crisis (SCD VOC). In a Phase 1 study of GMI-1070 infusion in SCD adults at steady state (not in VOC), GMI-1070 decreased markers of cellular activation including neutrophil integrins, platelet/neutrophil aggregates, soluble adhesion molecule concentration; and markers of hemostatic activation. Furthermore, in a randomized Phase 2 study of SCD patients, treatment of VOC with GMI-1070 improved clinical outcomes such as time to resolution of crisis, time to discharge, and IV opioid use. Herein we report on the effect of GMI-1070 on biomarkers of cellular and hemostatic cascade activation from this Phase 2 trial. Methods: Patients in VOC enrolled in a prospective, randomized multi-center double-blind Phase 2 trial, ages 12-60 with HbSS or HbSB0thalassemia were treated with GMI-1070 q12h or placebo, in addition to standard treatment per institutional practice, until resolution of VOC. Clinical outcomes and pharmacokinetics have been previously reported (ASH 2013 Abstracts 775, 776, and 2206). Biomarker blood samples were drawn prior to study drug, and on a sparse sampling basis at times starting 30 minutes after initial dose and continuing until 36 hours after the last dose. Analytes measured included: soluble adhesion molecules E-selectin (sEsel), P-selectin, L-selectin, intercellular adhesion molecules 1 and 3, vascular cell adhesion molecule-1; and tissue factor and thrombin-antithrombin complexes by ELISA. At some sites, surface expression of monocyte b2 integrins MAC-1 & LFA-1 and platelet-monocyte aggregates were also measured by flow cytometry. Comparisons were made between the GMI-1070 and placebo groups, and serial expression levels were compared over time. Subgroup analyses were performed by hydroxyurea (HU) use, age group, baseline WBC, and ‘responders' based on clinical outcomes. A mixed effect model was used to test the LS means difference at each time point and ANCOVA model was used to analyze the nadir, peak, and last dose values. Results: ELISA and flow cytometry samples were collected from 70 and 15 subjects, respectively. Soluble E-selectin levels were reduced for the group on GMI-1070 compared to placebo throughout hospitalization, and the differences were statistically significant at some time-points (Figure 1). Baseline sEsel levels were similar; but the peak, nadir, and level at last dose were all lower in the GMI-1070 group (Figure 2). Exploratory subgroup analysis by HU use, age group, response as measured by visual analog scale or opiate use, frequency of VOC in the past, and baseline white blood cell count revealed consistently lower sEsel levels in the GMI-1070 group. Many, but not all, of these differences reached statistical significance. Conclusion: GMI-1070 use during VOC resulted in consistent and significant reductions of sEsel, overall and in sub-groups as compared to placebo. These findings are consistent with the hypothesized effect of GMI-1070 on endothelial activation and/or apoptosis, mediated by inhibition of E-selectin, although an effect on sEsel clearance cannot be excluded. A Phase 3 study is planned to evaluate efficacy and safety of GMI-1070 as treatment for VOC. Soluble E-selectin concentrations may be useful as a biomarker of pharmacodynamic effect. Figure 1: sE-sel was reduced in the GMI-1070 group at all timepoints tested. Comparison to placebo for change from baseline over time is shown. *p<0.05 ***p<0.001 Figure 1:. sE-sel was reduced in the GMI-1070 group at all timepoints tested. Comparison to placebo for change from baseline over time is shown. *p<0.05 ***p<0.001 Figure 2: Figure 2: sE-sel was similar at baseline between GMI-1070 and placebo groups; mean change was different for GMI-1070 compared to placebo at the nadir, peak, and last dose values. Change from baseline is shown. *p<0.05 **p<0.01 Figure 2:. Figure 2: sE-sel was similar at baseline between GMI-1070 and placebo groups; mean change was different for GMI-1070 compared to placebo at the nadir, peak, and last dose values. Change from baseline is shown. *p<0.05 **p<0.01 Disclosures Wun: GlycoMimetics: Research Funding; Pfizer: Steering Committee, Steering Committee Other; Emmaus Medical: Consultancy. Telen:GlycoMimetics: Research Funding; Pfizer: Consultancy; Dilaforette: Research Funding. Krishnamurti:GlycoMimetics: Research Funding. McCavit:GlycoMimetics: Research Funding; Pfizer: Consultancy. DeCastro:GlycoMimetics: Research Funding. Flanner:GlycoMimetics: Employment, Equity Ownership. Kuypers:GlycoMimetics: Research Funding. Larkin:GlycoMimetics: Research Funding. Rhee:GlycoMimetics: Consultancy. Magnani:GlycoMimetics Inc.: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Thackray:GlycoMimetics: Employment, Equity Ownership.


2012 ◽  
Author(s):  
Mario García Molina ◽  
Liliana Chicaíza ◽  
Alexander Moreno Calderón ◽  
Víctor Prieto Martínez ◽  
Adriana Linares Ballesteros ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1772-1772
Author(s):  
Santiago Pavlovsky ◽  
Astrid Pavlovsky ◽  
Isolda Fernandez ◽  
Miguel Pavlovsky ◽  
Virginia Prates ◽  
...  

Abstract Abstract 1772 Background: Hodgkin Lymphoma (HL) is the most curable type of Lymphoma with an overall survival at 5 years of 80%. ABVD can be considered as gold standard for first line treatment for all stages of HL. Dividing patients (pts.) in different prognostic groups has aimed to reduce chemo and radio toxicity in those patients with good prognosis. A negative PET-CT, either early during treatment of ABVD or after completion of it, has shown to be a powerful prognostic tool (Hutchings: Blood 2006; Gallamini: Haematologica 2006). Our cooperative group has an experience with 584 patients with HL in early or advanced stage treated with 3 or 6 cycles of ABVD plus involved field radiotherapy with a complete remission (CR) of 91% and an event free survival (EFS) and overall survival (OS) at 60 months of 79% and 95%.(S Pavlovsky, Clin Lymp My & Leuk, 2010). Aims: Test the efficacy of treatment to all stages of HL adjusted to PET-CT results after 3 cycles of ABVD. Evaluate the outcome of pts. who have a negative PET-CT after 3 cycles of ABVD and receive no further treatment. Intensify therapy only in pts. who have persistent hyper metabolic lesions in PET-CT after 3 cycles of ABVD. Method: Since October 2005, 198 newly diagnosed pts. with HL have been included in a prospective multicenter trial. Initially all patients received 3 cycles of ABVD. After the third cycle, pts. were evaluated with a PET-CT. Those pts. who achieved CR with a negative PET-CT, received no further treatment. Those with more than 50% of anatomic reduction of initial masses but persistent hyper metabolic lesions by PET-TC after 3 ABVD were considered in partial remission (PR) and completed 6 cycles of ABVD and radiotherapy (RT) on PET-CT positive areas. Those patients with less than PR after 3 cycles of ABVD received ESHAP and if CR, high doses of chemotherapy and an autologous stem cell transplant (ASCT). All patients were re-evaluated at the end of treatment. The median follow up is of 30 months (3-62). Results: One hundred and seventy three patients completed three cycles of ABVD followed by a PET-CT. The median age at diagnosis was 29 years. One hundred and thirty-six (79%) had localized stage (I-II) at diagnosis and 37 (21%) presented with advanced stage (III-IV). Of 155 pts. 77 (50%) pts had IPS 0–1, 66 (43%) had IPS 2–3 and 12 (8%) had IPS 4–5. Twenty six (17%) pts. had bulky disease at diagnosis. One hundred and thirty-seven (79%) pts. achieved CR with negative PET-CT after 3 cycles of ABVD. Thirty-six (21%) were PET-CT positive, of them 32 pts achieved PR and completed a total of 6 cycles of ABVD plus RT in hyper metabolic lesions. Twenty five achieved CR (72%), 5 persisted with PR and 2 died of progressive disease. Four pts showed progressive disease (PD) after 3 ABVD and received ESHAP and ASCT, 2 achieved and remained in CR, 1 is in PR and 1 died of progressive disease. Of 173 pts who completed treatment with ABVD × 3 cycles, ABVD × 6 cycles plus RT on PET-TC positive areas or ESHAP plus ASCT, 164 pts (95%) achieved CR. Of these 164 pts., 14 pts (8%) relapsed. The EFS and OS at 36 months is 83% and 97% respectively. Patients with early negative PET-TC have an event-free survival of 87% compared to 62% (P=0,001) for pts with early positive PET CT. The OS at 36 months was 100% versus 86% respectively (<0.001). Conclusion: Treating patients with ABVD, evaluating response after 3 cycles with PET-CT, and adapting further therapy, leads to a high rate of CR avoiding more aggressive chemotherapy and radiotherapy. Three courses of ABVD without RT are adequate in patients with early CR defined by negative PET-CT. In early positive PET-CT it is possible to intensify therapy improving the otherwise bad prognosis; more aggressive treatment might also be suitable. These results need to be confirmed by a larger group of patients and a longer follow-up. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3644-3644 ◽  
Author(s):  
Lale Kostakoglu ◽  
Heiko Schoder ◽  
Nathan Hall ◽  
David J. Straus ◽  
Jeffrey L Johnson ◽  
...  

Abstract Abstract 3644 Use of interim PET in Hodgkin Lymphoma (HL) for risk-adapted treatment has been confounded by a lack of standardized criteria for interpretation. The International Harmonisation Project (IHP) criteria (JCO 2007;25:571) have been validated and are widely used for restaging following therapy. Our objective was to validate the IHP criteria for response evaluation based on PET after two cycles and correlate with the “London”criteria and diagnostic CT-based (dCT) lesion size changes. Methods: Pts were accrued prospectively to CALGB 50203, a trial of doxorubicin, vinblastine and gemcitabine (AVG) for initial treatment of stage I-II non-bulky Hodgkin lymphoma (HL). All had FDG PET or PET/CT and a separate high-resolution dCT scan at baseline, after two cycles of AVG (PET-2 and dCT-2) and at the end of therapy. No treatment change was made based on the PET-2 results. Of 99 assessable pts, 88 had both PET-2 and dCT-2. The primary PET-2 interpretation was based on IHP criteria (uptake > mediastinal blood pool/background is positive), a secondary interpretation was performed using the 5 point London criteria (uptake > liver is positive). The percent decrease in the sum of the products of the diameter (%SPPD) was determined between baseline dCT and dCT-2. A receiver operator curve (ROC) analysis was performed to determine the best cut-off for %SPPD to define a positive and negative CT result. The PET-2 and dCT-2 (%SPPD change) data were correlated with progression free survival (PFS). Results: Sixty-four pts (73%) achieved a complete remission (CR)/CR unconfirmed (CRu), 23.9% a partial response (PR), and 3% had stable disease. After a median follow-up of 3.3 years (1.8–5.0 years), 23.9% of patients relapsed/progressed with an estimated 3-year PFS of 0.77 [CI 68,84]. Eleven of 24 (45.8%) PET-2 positive patients relapsed vs. 10 of 64 (15.6%) PET-2 negative patients (p=0.0004). The best cut-off determined from ROC analysis for %SPPD change was 65%. The comparative results for individual evaluation criteria are displayed in the Table. In the PET-2 positive group, a negative dCT-2 increased PFS by 27–35%, suggesting an influence from dCT-2 results. However, in the combinatorial analysis, some of the confidence intervals are large due to small number of patients in each individual group, particularly, when both PET-2 and dCT-2 were positive as well as when PET-2 was positive and dCT-2 was negative. Conclusions: Interim PET/CT after two cycles of chemotherapy using either IHP or London criteria has a high NPV in early stage non-bulky HL. However, the PPV of interim PET needs to be improved to guide clinical management. Combining PET-2 with %SPPD decrease after 2 cycles improves prediction of PFS compared to each test alone. These data provide a proof of concept for risk-adapted clinical trials and further studies are underway to confirm these findings in a larger population. Disclosures: Bartlett: seattle genetics: Research Funding. Cheson:Celphalon: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celegen: Membership on an entity's Board of Directors or advisory committees, Research Funding; GlaxoSmithKline: Research Funding.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1538-1538
Author(s):  
Aristoteles Giagounidis ◽  
Alan List ◽  
Eva Hellström-Lindberg ◽  
Mikkael A. Sekeres ◽  
Ghulam J. Mufti ◽  
...  

Abstract Introduction The proportion of aberrant metaphases is prognostic for overall survival (OS) in MDS patients with trisomy 8 (Mallo M, et al. Leuk Res. 2011;35:834-6). The impact of the proportion of metaphases with del(5q) on clinical outcomes, including OS, disease progression and response to therapy with LEN in MDS remains undefined. In two large multicenter studies of LEN (MDS-003 and MDS-004) in RBC transfusion-dependent patients with International Prognostic Scoring System (IPSS) Low- or Intermediate (Int)-1-risk del(5q) MDS, RBC transfusion independence (TI) ≥ 8 weeks was achieved in 51–67% of patients (List A, et al. N Engl J Med. 2006;355:1456-65; Fenaux P, et al. Blood. 2011;118:3765-76). This retrospective analysis evaluated response to treatment, progression to acute myeloid leukemia (AML) and OS by proportion of del(5q) metaphases in patients with isolated del(5q) from the MDS-003 and 004 studies. Methods In order to allow sufficient patient numbers for analysis, ≥ 16 metaphases were evaluated for del(5q) by standard karyotyping (MDS-003 and MDS-004) and 200 interphase nuclei were evaluated by fluorescence in situ hybridization (FISH; MDS-004 only) using a probe for the commonly deleted region 5q31 (LSI EGR1/D5S721, Abbott, Wiesbaden, Germany). Patients received LEN on days 1–21 of each 28-day cycle (10 mg) or continuously (5 mg or 10 mg), or placebo. In MDS-004, patients randomized to placebo could cross over to LEN 5 mg by week 16. RBC-TI ≥ 26 weeks, time to AML progression and OS were analyzed by the proportion of del(5q) metaphases or interphases (≤ 60% vs > 60%) using standard karyotyping and FISH, respectively. Results Of the 353 patients from MDS-003 and MDS-004, 194 had isolated del(5q) by standard karyotyping; median proportion of del(5q) metaphases was 96% (range 4–100). Baseline characteristics including age, time from diagnosis, RBC transfusion burden, hemoglobin level, platelet and absolute neutrophil counts were comparable among patients with ≤ 60% (n = 21) and > 60% (n = 173) del(5q) metaphases. Rates of RBC-TI ≥ 26 weeks were similar across patients in the ≤ 60% and > 60% groups (P = 0.6515). Time to AML progression was comparable for patients in the ≤ 60% group versus the > 60% group (log-rank test P = 0.9802); 2-year rates were 22.2% (95% confidence interval [CI]: 7.7–54.5%) and 14.6% (95% CI: 9.9–21.2%), respectively. Time to AML progression was similar when analyzed with death without AML as competing risk (Gray’s test P = 0.5514). OS was longer in the > 60% versus the ≤ 60% group (log-rank test P = 0.0436); median OS was 3.7 years (95% CI: 3.0–4.2) and 2.4 years (95% CI: 1.5–4.9), respectively. In MDS-004, the proportion of del(5q) interphases was analyzed using FISH in 106 patients, including 46 with ≤ 60% and 60 with > 60%. When analyzed by FISH, rates of RBC-TI ≥ 26 weeks were similar across patients in the ≤ 60% and > 60% groups (P = 1.000). Time to AML progression and OS were similar across these groups (log-rank test P = 0.7311 and P = 0.8639, respectively) when analyzed by FISH. In the ≤ 60% and > 60% groups respectively, 2-year AML progression rates were 14.8% (95% CI: 6.9–30.1%) and 18.6% (95% CI: 10.4–32.0%), and median OS was 3.1 years (95% CI: 2.3–4.8) and 2.9 years (95% CI: 2.3–4.4). Time to AML progression was similar when analyzed with death without AML as competing risk (Gray’s test P = 0.8631). Conclusions In IPSS Low- or Int-1-risk MDS patients with isolated del(5q) treated with LEN in MDS-003 and MDS-004 studies, baseline characteristics, RBC-TI ≥ 26 weeks and AML progression were comparable in patients with > 60% versus ≤ 60% del(5q) metaphases. Although similar across groups when analyzed by FISH in a subset of patients, surprisingly, OS was longer in patients with > 60% del(5q) metaphases than in those with ≤ 60% del(5q) metaphases by standard karyotyping. However, the number of patients with ≤ 60% del(5q) metaphases was limited and no adjustment was made for multiple testing. These findings suggest that the number of cells with the isolated del(5q) abnormality measured by FISH does not impact clinical outcome in this RBC transfusion-dependent study population, but this finding could not be confirmed for OS by standard karyotyping. Disclosures: Giagounidis: Celgene: Consultancy, Honoraria, Membership on an entity’s Board of Directors or advisory committees. List:Celgene: Serve on Celgene Data Safety & Monitoring Committee Other. Hellström-Lindberg:Celgene: Membership on an entity’s Board of Directors or advisory committees, Research Funding. Sekeres:Celgene: Membership on an entity’s Board of Directors or advisory committees; Amgen: Membership on an entity’s Board of Directors or advisory committees. Mufti:Celgene: Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding. Schlegelberger:Celgene: Consultancy. Morrill:Celgene: Employment, Equity Ownership. Wu:Celgene: Employment, Equity Ownership. Skikne:Celgene: Employment, Equity Ownership. Fenaux:Celgene: Honoraria.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 292-292 ◽  
Author(s):  
Joseph M Connors ◽  
Stephen Ansell ◽  
Steven I. Park ◽  
Michelle A. Fanale ◽  
Anas Younes

Background: The ABVD regimen containing doxorubicin, bleomycin, vinblastine, and dacarbazine is a common standard of care for the frontline treatment of advanced stage Hodgkin lymphoma (Santoro 1987; Duggan 2003) and is curative for the majority of patients; however, up to 30% of patients require a secondary therapy. Hodgkin Reed-Sternberg cells of classical HL (cHL) typically express CD30. In a pivotal phase 2 trial brentuximab vedotin (A, ADCETRIS®), comprised of an anti-CD30 monoclonal antibody conjugated by a protease-cleavable linker to a microtubule-disrupting agent, monomethyl auristatin E (MMAE) induced an objective response rate (ORR) of 75% and complete response rate (CR) of 34% in highly treatment-refractory patients with cHL (Younes 2012). Methods: We conducted a phase 1, open-label, multicenter study to evaluate the safety and efficacy of A when administered in combination with standard therapy (ABVD) or the same regimen without bleomycin (AVD) (Younes 2013). Adult patients with newly diagnosed advanced stage (II bulky, II B, III or IV; 80% stage III or IV) received doses of 0.6, 0.9, or 1.2 mg/kg A with standard doses of ABVD or 1.2 mg/kg with AVD, depending on cohort assignment on Days 1 and 15 of each 28-day cycle for up to 6 cycles of therapy. Previously we reported that among patients assessable for response 95% of patients given ABVD+A achieved a CR, as did 96% of patients given AVD+A. None of 26 patients given AVD+A but 11 of 25 (44%) given ABVD+A experienced pulmonary toxicity, including 2 deaths, establishing that A cannot be safely combined with bleomycin. In this current study we provide the long term survival and safety data on patients enrolled in the phase 1 trial. Results: In total 51 patients were assigned to either ABVD+A (n=25) or AVD+A (n=26). 1 patient who withdrew from the trial during the first cycle of ABVD+A is excluded from this analysis and 1 patient who received 3 cycles of ABVD+A, then withdrew, then received 3 cycles of ABVD alone and 2 patients who died during treatment (pulmonary toxicity) are included (total n=50). Median follow-up from diagnosis for the 24 patients treated with ABVD+A is 41 months (range 9-51 months) and for the 26 patients treated with AVD+A, 31 months (range 9-35 months). All 26 patients treated with AVD+A have been followed longer than the longest time to relapse (7 months). 45 patients remain in first CR and 5 treatment failures have occurred: 4 in the ABVD+A cohort (2 toxic deaths; 2 relapses (9 and 23 months from diagnosis)) and 1 after AVD+A (7 months from diagnosis). 3y-failure-free survival (3y-FFS) is 83% and 96% for ABVD+A and AVD+A, respectively, and 3y-overall survival (3y-OS), is 92% and 100%. No additional toxic deaths have occurred in follow-up. Conclusions: These updated outcomes reflecting the impact of adding brentuximab vedotin (1.2 mg/kg) to standard doses of AVD for classical Hodgkin lymphoma, demonstrating 96% 3y-FFS and 100% 3y-OS with no major unexpected toxicity, strongly support the current large international trial comparing AVD-A (AVD+1.2mg/kg brentuximab vedotin) to standard ABVD (ECHELON-1, clinicaltrials.gov NCT01712490), which may identify a new, less toxic gold standard treatment for advanced stage classical Hodgkin lymphoma. Disclosures Connors: Seattle Genetics: Research Funding. Off Label Use: brentuximab vedotin in phase 1 trial. Ansell:Seattle Genetics: Research Funding. Park:Seattle Genetics: Research Funding; Millennium/Takeda: Research Funding. Fanale:Seattle Genetics: Research Funding. Younes:Seattle Genetics: Research Funding; Millennium/Takeda: Research Funding.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2656-2656
Author(s):  
Steven R. Arikian ◽  
Dejan Milentijevic ◽  
Gary Binder ◽  
Mara Silvia Monzini ◽  
X Henry Hu ◽  
...  

Abstract Introduction: As clinical evidence has mounted in support of novel agents and longer treatment (Tx) durations for patients (pts) with newly diagnosed multiple myeloma (NDMM), questions have arisen regarding the economic impact of extending time to progression (TTP) in these pts, and the cost consequences once pts relapse and move to a second line of Tx. Previous analysis showed that relapsed myeloma pts incurred higher monthly costs once they advanced to later lines of Tx (Gaultney, 2013). There is limited information on the cost patterns of MM pts before and after their first relapse. A claims analysis was performed to evaluate the patterns of total direct costs of care, from Tx initiation until progression, for NDMM patients and for newly relapsed patients treated with novel agents, utilizing time to next therapy (TTNT) as a proxy measure for progression. Methods: A retrospective study was conducted using a large US medical and pharmacy claims database, covering > 25 million lives annually. NDMM patients were identified with at least 2 outpatient claims or 1 inpatient medical claim associated with a diagnosis of MM (ICD-9-CM] code 203.0X), with the first such claim used to define the index date. Inclusion criteria required a minimum of 12 months' pre-index enrollment and 6 months' post-index continuous enrollment between 2006 and 2012. Pts with claims for stem cell transplantation (SCT) were excluded, to avoid confounding results from various factors based on timing, costs, and site of care of SCT. The analysis focused on NDMM and relapsed MM pts receiving lenalidomide (LEN)- or bortezomib (BORT)- based Tx, where complete claim history was available from Tx onset to initiation of subsequent Tx. Using methods similar to those described by Gaultney, patients' average monthly costs were determined, including medical (inpatient, ambulatory, and emergency room) and pharmacy (index and other drugs) costs, and total cost patterns over quarterly time periods were calculated. Average Charlson comorbidity scores were determined to compare baseline measures between pt groups. Results: 897 NDMM pts and 280 relapsed MM pts were identified with complete data through initiation of subsequent Tx. Monthly total direct costs for NDMM pts were $15,400 in the first 3 months (mos) of Tx, and declined each quarter, reaching approximately $5,000/mo at 18+ mos. At relapse, monthly costs increased to over $12,000 for the first 3 mos and followed a quarterly pattern of reduction similar to that seen for NDMM pts (Fig 1). Quarterly cost reduction patterns were consistent for patients treated with LEN or BORT for both NDMM and relapsed pts. Pts' total monthly NDMM costs over the full TTNT period averaged $8,942 with LEN vs. $11,139 for BORT (due to 54% higher monthly medical costs for BORT), while monthly drug costs were nearly identical (Table 1). The baseline Charlson comorbidity index was similar between Tx groups in both lines of Tx. Figure 1: Direct monthly costs (medical and pharmacy) for LEN- and BORT-based treatments Figure 1:. Direct monthly costs (medical and pharmacy) for LEN- and BORT-based treatments Table 1: Direct monthly costs for NDMM pts Table 1 Table 1. Conclusions: For a population of NDMM pts receiving either LEN- or BORT-based Tx without SCT, followed until TTNT, total direct monthly costs per pt declined steadily over time, decreasing by 68% from the initial quarter to the period post 18 mos. Costs spiked when pts began 2nd-line therapy, then followed a similar pattern of decline over time. This pattern may suggest that further extending the TTP for NDMM pts may also yield economic benefits for each month extended before relapse. Patterns of cost decline were similar between the LEN and BORT groups, for NDMM and for relapsed patients, although mean monthly total costs were lower for NDMM pts receiving LEN-based Tx due to lower medical costs and similar drug costs. Disclosures Arikian: Genesis Research: Consultancy. Off Label Use: Lenalidomide in newly diagnosed multiple myeloma patients . Milentijevic:Celgene Corporation: Consultancy. Binder:Celgene Corporation: Employment, Equity Ownership. Monzini:Celgene Corporation: Employment, Equity Ownership. Hu:Celgene Corporation: Employment. Nagarwala:Celgene Corporation: Employment. Hussein:Celgene Corporation: Employment. Corvino:Genesis Research LLC: Consultancy. Surinach:Genesis Research LLC: Consultancy. Usmani:Celgene Corporation: Consultancy, Honoraria, Research Funding; Millennium: Consultancy, Honoraria; Onyx: Consultancy, Honoraria, Research Funding; Sanofi: Consultancy; Array BioPharma: Research Funding; Janssen: Research Funding; Pharmacyclics: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document