8 research outputs found
Daratumumab in first-line treatment of patients with light chain amyloidosis and Mayo stage IIIb improves treatment response and overall survival
Treatment of patients with Mayo stage IIIb light chain (AL) amyloidosis is still challenging, and the prognosis remains very poor. Mayo stage IIIb patients were excluded from the pivotal trial leading to the approval of daratumumab in combination with bortezomib-cyclophosphamide-dexamethasone. This retrospective, multicenter study evaluates the addition of daratumumab to first-line therapy in patients with newly diagnosed stage IIIb AL amyloidosis. In total, data from 119 consecutive patients were analyzed, 27 patients received an upfront treatment including daratumumab, 63 a bortezomibbased regimen without daratumumab, eight received therapies other than daratumumab or bortezomib and 21 pretreated patients or deceased prior to treatment were excluded. In the daratumumab group, median overall survival was not reached after a median follow-up time of 14.5 months, while it was significantly worse in the bortezomib- and the otherwise treated group (6.6 and 2.2 months, respectively) (P=0.002). Overall hematologic response rate at 2 and 6 months was better in the daratumumab group compared to the bortezomib group (59% vs. 37%, P=0.12, 67% vs. 41%, P=0.04, respectively). Landmark survival analyses revealed a significantly improved overall survival in patients with partial hematologic response or better, compared to non-responders. Cardiac response at 6 months was 46%, 21%, 0% in the daratumumab-, bortezomib- and otherwise treated groups, respectively (P=0.04). A landmark survival analysis revealed markedly improved overall survival in patients with cardiac very good partial response vs. cardiac non-responders (P=0.002). This study demonstrates for the first time the superiority of an upfront treatment with daratumumab over standard-of-care in stage IIIb AL amyloidosis
Elotuzumab, pomalidomide, and dexamethasone is a very well tolerated regimen associated with durable remission even in very advanced myeloma: a retrospective study from two academic centers
Background
The anti-SLAMF7 monoclonal antibody, elotuzumab (elo), plus lenalidomide (len) and dexamethasone (dex) is approved for relapsed/refractory MM in the U.S. and Europe. Recently, a small phase 2 study demonstrated an advantage in progression-free survival (PFS) for elo plus pomalidomide (pom)/dex compared to pom/dex alone and resulted in licensing of this novel triplet combination, but clinical experience is still limited.
Purpose
To analyze the efficacy and safety of elo/pom/dex in a “real world” cohort of patients with advanced MM, we queried the databases of the university hospitals of Würzburg and Vienna.
Findings
We identified 22 patients with a median number of five prior lines of therapy who received elo/pom/dex prior to licensing within an early access program. Patients received a median number of 5 four-week treatment cycles. Median PFS was 6.4 months with 12-month and 18-month PFS rates of 35% and 28%, respectively. The overall response rate was 50% and 64% of responding patients who achieved a longer PFS with elo/pom/dex compared to their most recent line of therapy. Objective responses were also seen in five patients who had been pretreated with pomalidomide. Low tumor burden was associated with improved PFS (13.5 months for patients with ISS stage I/II at study entry v 6.4 months for ISS III), although this difference did not reach statistical significance. No infusion-related reactions were reported. The most frequent grade 3/4 adverse events were neutropenia and pneumonia.
Conclusion
Elo/pom/dex is an active and well-tolerated regimen in highly advanced MM even after pretreatment with pomalidomide
Elotuzumab, pomalidomide, and dexamethasone is a very well tolerated regimen associated with durable remission even in very advanced myeloma: a retrospective study from two academic centers
Abstract
Background
The anti-SLAMF7 monoclonal antibody, elotuzumab (elo), plus lenalidomide (len) and dexamethasone (dex) is approved for relapsed/refractory MM in the U.S. and Europe. Recently, a small phase 2 study demonstrated an advantage in progression-free survival (PFS) for elo plus pomalidomide (pom)/dex compared to pom/dex alone and resulted in licensing of this novel triplet combination, but clinical experience is still limited.
Purpose
To analyze the efficacy and safety of elo/pom/dex in a “real world” cohort of patients with advanced MM, we queried the databases of the university hospitals of Würzburg and Vienna.
Findings
We identified 22 patients with a median number of five prior lines of therapy who received elo/pom/dex prior to licensing within an early access program. Patients received a median number of 5 four-week treatment cycles. Median PFS was 6.4 months with 12-month and 18-month PFS rates of 35% and 28%, respectively. The overall response rate was 50% and 64% of responding patients who achieved a longer PFS with elo/pom/dex compared to their most recent line of therapy. Objective responses were also seen in five patients who had been pretreated with pomalidomide. Low tumor burden was associated with improved PFS (13.5 months for patients with ISS stage I/II at study entry v 6.4 months for ISS III), although this difference did not reach statistical significance. No infusion-related reactions were reported. The most frequent grade 3/4 adverse events were neutropenia and pneumonia.
Conclusion
Elo/pom/dex is an active and well-tolerated regimen in highly advanced MM even after pretreatment with pomalidomide.
</jats:sec
Open-Label, Prospective, Multicentre, Phase I/II Study of AOP2014, a Novel PEG-Proline-Interferon Alpha-2b in Patients with Polycythemia Vera: Update from an Ongoing Study
Abstract
Abstract 1747
Background. IFN-alpha (IFN) is reported to induce molecular response in patients with chronic myeloproliferative neoplasms and may therefore improve survival. However, the use of conventional and pegylated IFNs is limited by toxicity, leading to treatment withdrawals in a substantial proportion of patients. In polycythemia vera (PV), recently published phase II data indicate occurence of grade 3 and 4 toxicities in 20%, and toxicity-related discontinuations in 22–24% of patients administered once weekly with pegylated IFNs. Results from large-scale randomized studies of IFNs in PV are not available. AOP2014 is a next generation long-acting IFNa-2b, consisting predominantly of only one isoform as opposed to other commercially available pegylated interferons. AOP2014 is expected to have pharmacokinetic and pharmacodynamic profiles which may allow a reduced dosing frequency compared to other pegylated IFNs, potentially leading to improved tolerability, better compliance, and, finally, favourable long-term treatment outcomes.
Study design. This is an open-label, prospective, multicentre, phase I/II dose escalation study of AOP2014 to determine the maximum tolerated dose (MTD), safety and efficacy in PV. Inclusion criteria are: age ≥18 years, confirmed diagnosis of PV (WHO, 2008, or the PSVG plus JAK2 positivity). Both newly diagnosed and pre-treated (cytoreduction) patients are eligible; hydroxyurea (HU) is allowed until the first application of AOP2014. If abnormally elevated prior to entry, sustained hematocrit (Hct) ≤45% is to be achieved by phlebotomy prior to the first administration. 3+3 dose escalation with cohort extension after MTD definition is being utilized. Intra-patient dose escalation to the dose level, previously established to be safe, is encouraged. MTD is defined as the highest dose level at which no more than 1 of 6 subjects experience a DLT during the first treatment period (2 weeks). Complete hematological response (CR) is defined by Hct <45%, platelet count ≤400*109/L, WBC count ≤10*109/L, normal spleen size, and absence of thromboembolic events. Partial response (PR) is defined as haematocrit <45% without phlebotomy but with persistent splenomegaly or elevated (>400*109/L) platelet count, or reduction of phlebotomy requirements by at least 50%. Evaluation of toxicities and JAK2 allele burden quantication will be performed.
Results. 20 patients have been included to date, M/F ratio was 16/4, median age 58 years (range 44–83). Median time from diagnosis to inclusion was 19 months (range 0–170). 9 patients were pre-treated with HU prior to study entry. Median number of phlebotomies in the past 3 months prior to inclusion was 1 (range 0–4), a total of 13 patients were regularly phlebotomized at least once in three months prior to study entry. Six patients had a history of thrombotic complications. Median Hct at baseline was 46% in males (range 46–52) and 42% in females (range 41–43). Median WBC and platelet counts were 11.3*109/L (range 4.7–17.8) and 366*109/L (range 148–1016), respectively. Twelve patients (60%) had splenomegaly. Dose levels of 50, 100, 150, 225, 300, 360 and 450 microgramm once in two weeks were investigated. The median reported treatment duration is 24 weeks (range 0–33). After 10 weeks of treatment (14 evaluable patients), 64% of patients had hematological response (2 CR; 7 PR), one patient needed to be phlebotomized. At 18 weeks (9 evaluable patients), 1 patient had CR and 4 – PR, overall response (CR+PR) was 56%; at 28 weeks (8 evaluable patients), 1 patient had CR and 5 PR, overall response was 63%. The only evaluable patient at week 34 had CR. Starting from week 18, no patient required phlebotomy. Mainly grade 1 and 2 adverse events were reported. Administration site reactions developed in 11 patients, all of mild intensity and recovered within 72h. The most frequent adverse reactions were muscle and joint pain and gastrointestinal toxicities (12 and 6 patients, respectively). Fever and depression were observed in 5 and 2 patients, respectively. Drug-related adverse event (depression) was the reason of treatment discontinuation in one patient. Molecular response data are still undergoing evaluation.
Conclusion. Response rate of above 60% at six month already observed in the phase I dose finding part suggests that AOP2014 has promising efficacy and good tolerability profile in PV. Updated results will be presented as the trial progresses.
Disclosures:
Gisslinger: AOP Orphan Pharmaceuticals AG: Research Funding, Speakers Bureau; Novartis: Speakers Bureau; Celgene Austria: Research Funding, Speakers Bureau. Kralovics:(7) AOP Orphan Pharmaceuticals AG: Research Funding. Schoder:AOP Orphan Pharmaceuticals AG: Research Funding. Gisslinger:AOP Orphan Pharmaceuticals AG: Research Funding. Buxhofer-Ausch:AOP Orphan Pharmaceuticals AG: Research Funding. Strecker:AOP Orphan Pharmaceuticals AG: Research Funding. Wolf:AOP Orphan Pharmaceuticals AG: Research Funding. Willenbacher:AOP Orphan Pharmaceuticals AG: Research Funding. Greil:AOP Orphan Pharmaceuticals AG: Research Funding. Egle:AOP Orphan Pharmaceuticals AG: Research Funding. Melchardt:AOP Orphan Pharmaceuticals AG: Research Funding. Burgstaller:AOP Orphan Pharmaceuticals AG: Research Funding. Tarmann:AOP Orphan Pharmaceuticals AG: Employment. Wachter:AOP Orphan Pharmaceuticals AG: Employment. Zahriychuk:AOP Orphan Pharmaceuticals AG: Employment. Thaler:AOP Orphan Pharmaceuticals AG: Research Funding.
</jats:sec
Open-Label, Prospective, Multicentre, Phase I/II Study of AOP2014, a Novel PEG-Proline-Interferon Alpha-2b in Patients with Polycythemia Vera: Update from an Ongoing Study
EBioMedicine / Extracorporeal IgE Immunoadsorption in Allergic Asthma: Safety and Efficacy
Background
Prevention of IgE-binding to cellular IgE-receptors by anti-IgE (Omalizumab) is clinically effective in allergic asthma, but limited by IgE threshold-levels. To overcome this limitation, we developed a single-use IgE immunoadsorber column (IgEnio). IgEnio is based on a recombinant, IgE-specific antibody fragment and can be used for the specific extracorporeal desorption of IgE.
Objective
To study safety and efficacy of IgEnio regarding the selective depletion of IgE in a randomized, open-label, controlled pilot trial in patients with allergic asthma and to investigate if IgEnio can bind IgE-Omalizumab immune complexes.
Methods
Fifteen subjects were enrolled and randomly assigned to the treatment group (n = 10) or to the control group (n = 5). Immunoadsorption was done by veno-venous approach, processing the twofold calculated plasma volume during each treatment. A minimum average IgE-depletion of 50% after the last cycle in the intention-to-treat population was defined as primary endpoint. Safety of the treatment was studied as secondary endpoint. In addition, possible changes in allergen-specific sensitivity were investigated, as well as clinical effects by peak flow measurement and symptom-recording. The depletion of IgE-Omalizumab immune complexes was studied in vitro.
The study was registered at clinicaltrials.gov (NCT02096237) and conducted from December 2013 to July 2014.
Results
IgE immunoadsorption with IgEnio selectively depleted 86.2% (5.1% SD) of IgE until the end of the last cycle (p < 0.0001). Removal of pollen allergen-specific IgE was associated with a reduction of allergen-specific basophil-sensitivity and prevented increases of allergen-specific skin-sensitivity and clinical symptoms during pollen seasons. IgEnio also depleted IgE-Omalizumab immune complexes in vitro.
The therapy under investigation was safe and well-tolerated. During a total of 81 aphereses, 2 severe adverse events (SAE) were recorded, one of which, an episode of acute dyspnea, possibly was related to the treatment and resolved after administration of antihistamines and corticosteroids.
Conclusions
This pilot study indicates that IgE immunoadsorption with IgEnio may be used to treat patients with pollen-induced allergic asthma. Furthermore, the treatment could render allergic patients with highly elevated IgE-levels eligible for the administration of Omalizumab and facilitate the desorption of IgE-Omalizumab complexes.(VLID)485282
