Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Journal of Medicinal Chemistry, American Chemical Society (ACS), Vol. 63, No. 20 ( 2020-10-22), p. 11639-11662
    Type of Medium: Online Resource
    ISSN: 0022-2623 , 1520-4804
    Language: English
    Publisher: American Chemical Society (ACS)
    Publication Date: 2020
    detail.hit.zdb_id: 1491411-6
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Rapid Communications in Mass Spectrometry, Wiley, Vol. 37, No. 7 ( 2023-04-15)
    Abstract: Volatile organic compounds (VOCs) emitted by an artificial leather part for car interiors are determined using GC–MS (gas chromatograph coupled to a mass spectrometer) using simultaneous electron and chemical ionization (EI & CI). A device for swift reagent ion switching in CI mode between consecutive runs is presented. Methods VOCs emitted from the investigated material were sampled onto Tenax® absorption tubes using micro emission chambers and subsequently injected into the GC through thermal desorption. The detector was a time‐of‐flight mass spectrometer (TOFMS) simultaneously operating in EI and CI modes during a single chromatographic run. A custom permeation tube setup allowed for swift selection between various reagent ions in CI mode, e.g., [N 2 H] + , [H 3 O] + , [(H 2 O) 2 H] + , and [NH 4 ] + . Results Different reagent ions are swiftly selectable between single GC runs without hardware changes. Differences in precursor ion survival yields and the selectivity of the various reactants were carefully assessed. Several examples for the improved identification of unknown compounds with the available complementary and comprehensive EI & CI data set are demonstrated for a relevant material emission application. Conclusion The presented technique provides additional value to the standard GC‐EI/MS procedure commonly used for material emission characterization. It allows for a non‐targeted analysis approach with moderate analysis time.
    Type of Medium: Online Resource
    ISSN: 0951-4198 , 1097-0231
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2023
    detail.hit.zdb_id: 2002158-6
    detail.hit.zdb_id: 58731-X
    SSG: 11
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: European Urology, Elsevier BV, Vol. 48, No. 6 ( 2005-12), p. 922-931
    Type of Medium: Online Resource
    ISSN: 0302-2838
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2005
    detail.hit.zdb_id: 1482253-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Plant Biotechnology Journal, Wiley, Vol. 21, No. 6 ( 2023-06), p. 1240-1253
    Abstract: Rapid adaptation of weeds to herbicide applications in agriculture through resistance development is a widespread phenomenon. In particular, the grass Alopecurus myosuroides is an extremely problematic weed in cereal crops with the potential to manifest resistance in only a few generations. Target‐site resistances (TSRs), with their strong phenotypic response, play an important role in this rapid adaptive response. Recently, using PacBio's long‐read amplicon sequencing technology in hundreds of individuals, we were able to decipher the genomic context in which TSR mutations occur. However, sequencing individual amplicons are costly and time‐consuming, thus impractical to implement for other resistance loci or applications. Alternatively, pool‐based approaches overcome these limitations and provide reliable allele frequencies, although at the expense of not preserving haplotype information. In this proof‐of‐concept study, we sequenced with PacBio High Fidelity (HiFi) reads long‐range amplicons (13.2 kb), encompassing the entire ACCase gene in pools of over 100 individuals, and resolved them into haplotypes using the clustering algorithm PacBio amplicon analysis ( pbaa ), a new application for pools in plants and other organisms. From these amplicon pools, we were able to recover most haplotypes from previously sequenced individuals of the same population. In addition, we analysed new pools from a Germany‐wide collection of A. myosuroides populations and found that TSR mutations originating from soft sweeps of independent origin were common. Forward‐in‐time simulations indicate that TSR haplotypes will persist for decades even at relatively low frequencies and without selection, highlighting the importance of accurate measurement of TSR haplotype prevalence for weed management.
    Type of Medium: Online Resource
    ISSN: 1467-7644 , 1467-7652
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2023
    detail.hit.zdb_id: 2136367-5
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Haematologica, Ferrata Storti Foundation (Haematologica), Vol. 104, No. 11 ( 2019-11), p. 2265-2273
    Type of Medium: Online Resource
    ISSN: 0390-6078 , 1592-8721
    Language: English
    Publisher: Ferrata Storti Foundation (Haematologica)
    Publication Date: 2019
    detail.hit.zdb_id: 2186022-1
    detail.hit.zdb_id: 2030158-3
    detail.hit.zdb_id: 2805244-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 404-404
    Abstract: We investigated if bortezomib during induction and maintenance improves survival in newly diagnosed Multiple Myeloma (MM). Methods 827 eligible patients with newly diagnosed symptomatic MM were randomized to receive induction therapy with VAD (vincristine, doxorubicin, dexamethasone; n=414) or PAD (bortezomib, doxorubicin, dexamethasone; n=413) followed by high-dose melphalan (HDM) and autologous stem cell transplant (ASCT). Maintenance consisted of daily thalidomide 50 mg (VAD) or 2-weekly bortezomib 1.3 mg/m2 i.v. (PAD) for 2 years. The primary analysis was progression-free survival (PFS) adjusted for ISS stage. We here report long-term results of this trial (P. Sonneveld et al., J Clin Oncol 2012;30:2946-2955). Results The number of eligible patients, patient characteristics and disease characteristics are similar to those reported before. The response rates during protocol treatment have improved slightly since all patients have now completed treatment: CR+nCR 49% vs 35%, VGPR 26% vs 21% and ≥PR 91% vs 83% (PAD vs VAD). After a median follow-up of 67 months, 111 of patients treated with VAD and 131 of patients treated with PAD were progression-free and alive. Progression-free survival (PFS) defined as time from randomization until progression, relapse or death (censored at date of alloSCT, if applicable), was superior with PAD when adjusted for ISS, (HR=0.78, 95% CI [0.66-0.91], P=.002) and in multivariate analysis (HR=0.76 (95% CI [0.64-0.90] , P=.001). For the secondary endpoint overall survival (OS) the PAD arm was superior when adjusted for ISS (HR=0.80, 95% CI [0.65-1.00], P=.047) as well as in multivariate analysis (HR=0.78, 95% CI [0.63-0.97] , P=.027). Landmark analysis from start of maintenance for PFS did not show a significant difference between Thalidomide and Bortezomib maintenance, however, for OS the PAD arm was superior (P=.035) (HR=0.71, 95% CI [0.52-0.98]). Subgroup analysis performed on patients with renal failure at presentation (serum creatinine ≥2 mg/dL; 45 VAD, 36 PAD) showed that the PAD arm was significantly superior for PFS (HR=0.44, 95% CI [0.26-0.75] , P=.003) and OS (HR=0.38, 95% CI [0.21-0.69], P 〈 .001). 35/45 (78%) patients treated with VAD had died as compared with 16/36 (44%) patients with PAD. In bortezomib treated patients with serum creatinine ≥2 mg/dL OS and PFS were not significantly different from those in bortezomib-treated patients with normal renal function. Analysis for the effect of study group, concentrating on the difference of single HDM (sHDM, HOVON policy) vs double HDM (dHDM, GMMG policy) with ASCT indicated that dHDM was not superior across treatment arms for PFS, but in multivariate analysis remained superior for OS (HR=0-72, 95% CI [0.58-0.90], P=.004). Comparison of study group and treatment arms for OS at 5 years indicated that PAD plus dHDM was superior (72% vs 59% (PAD+sHDM) vs 63% (VAD+dHDM) vs 56% (VAD+sHDM), logrank P=.004. When patients were classified by prognostic characterization by FISH and ISS (K. Neben et al. Blood 2012;119:940-948) subgroup analysis in patients treated with dHDM showed that in the VAD arm, PFS was significantly lower in intermediate risk (IRi) (HR 2.26, 95% CI [1.42-3.58] , P 〈 0.001) and poor risk (PRi) (HR 5.02, 95% CI [2.89-8.73], P 〈 0.001) as compared to good risk (GRi), while in the PAD arm the HR for PRi was 2.04, 95% CI [1.20-3.48], P=0.009, and for IRi 1.26, 95% CI [0.82-1.92] , P=0.29. These data suggest that with PAD+dHDM only high-risk remains an independent unfavourable group for PFS. For OS a comparable difference was observed. Finally, the actuarial probability to develop a second primary malignancy was not different between treatment arms (PAD 3% at 5 years vs VAD (5%), P=.22). Conclusions Bortezomib during induction and maintenance improves CR and achieves superior PFS and OS. Subgroup analysis after long follow-up confirms the favourable outcome in patients with renal failure. Comparison of study subgroup analysis suggests that bortezomib treatment combined with double intensive treatment may be beneficial for PFS/OS. This trial has been registered as NTR213; EudraCT no. 2004-000944-26; ISRCTN: 64455289. This trial was supported by the Dutch Cancer Foundation, the German Federal Ministry of Education and Research and an unrestricted grant from Janssen-Cilag-Ortho Biotech. The GMMG study group received further grants to support this trial by Novartis, AMGEN, Chugai and Roche. Disclosures: Sonneveld: Janssen-Cilag: Honoraria; Celgene: Honoraria; Onyx: Honoraria; Janssen-Cilag: Research Funding; Millenium: Research Funding; Onyx: Research Funding; Celgene: Research Funding. Salwender:janssen: Honoraria. Weisel:Celgene: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria. Schmidt-Wolf:janssen: Research Funding; novartis: Honoraria. van de Velde:Janssen Research & Development: Employment; Johnson & Johnson: Equity Ownership. Lokhorst:Genmab A/S: Consultancy, Research Funding; Celgene: Honoraria; Johnson-Cilag: Honoraria; Mudipharma: Honoraria. Goldschmidt:janssen: Honoraria, Research Funding; amgen: Research Funding; novartis: Honoraria, Research Funding; chigai: Honoraria, Research Funding; roche: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 633-633
    Abstract: Abstract 633 Introduction: This independent phase 2 trial was designed to evaluate carfilzomib (C) combined with thalidomide and dexamethasone during induction and consolidation for feasibility, response and progression-free survival (PFS) in patients with newly diagnosed symptomatic MM, who were candidates for high-dose therapy. Patients with symptomatic MM and measurable disease, age 15 to 65 and no significant co-morbidity were eligible. At diagnosis Fluorescent in situ Hybridization (FISH) was performed of recurrent translocations, trisomy 9, del(17p), del (13q) and add(1q) Patients received 4 cycles of carfilzomib at 20 mg/m2 on days 1 & 2 followed by 27mg/m2 on days 8,9,15,16 of cycle 1 and on days 1,2, 8, 9, 15 & 16 of all subsequent 28-day cycles, thalidomide 200 mg days 1 – 28 of a 28 day cycle and dexamethasone 40 mg days 1, 8, 15 & 22 of a 28 day cycle. Stem cell harvest was performed with cyclophosphamide 2 g/m2 and G-CSF. Following HDM (200 mg/m2) and autologous stem cell transplantation (ASCT), consolidation therapy consisted of 4 cycles of carfilzomib 27 mg/m2 days 1, 2, 8, 9, 15 & 16 of a 28 day cycle, thalidomide 50 mg days 1–28 of a 28 day cycle and dexamethasone 20 mg days 1, 8, 15, 22 of a 28 day cycle. The primary endpoint was response, other endpoints were complete response (CR) according to IMWG criteria, immunofixation-negative CR (sCR), VGPR all pre-and post HDM, PFS and overall survival (OS). An interim analysis was planned after 20 evaluable patients, primarily to guard against excessive toxicity and/or lack of response. Results: While recruitment is still ongoing, 34 patients have been included, of which the first 20 patients were are evaluated for response and toxicity, with a median follow-up of 5 months. One patient was excluded because unavailability of data. Median age was 60 yr and ISS stages I/II/III were 8/6/5, respectively. Four patients went off treatment because of intolerance to thalidomide (n=1), tumor lysis syndrome with renal failure (n=1) or respiratory infections (n=2). Adverse events CTC grade 3+4 included tumor lysis syndrome (n=2), metabolic disorders (n=4), cardiovascular including DVT (n=5), gastrointestinal (n=2), skin rash (n=2) and reversible renal failure (n=3). Peripheral polyneuropathy grades 1+ 2 was observed in 7 (35%) of patients, but no grade 3 or higher. Responses after cycle 1 were CR + sCR 5%, VGPR 32%, PR 47%, SD 10%, NE 5% and after induction overall CR + sCR 21%, VGPR 47%, PR 16%, SD 10%, NE 5%. Median time to maximum response was 1 cycle. Secondary analysis revealed that responses occurred across cytogenetic subgroups as determined by FISH, i.e. add (1q) (n=2), t(4;14) (n=2), del(17p) (n=1) and del(13q) (n=5). Stem cell harvest was accomplished with standard CD34+ yield in all patients and HDM/ASCT was performed with complete hematologic recovery in 4/4 patients. Conclusion: Carfilzomib combined with thalidomide and dexamethasone during induction and consolidation is feasible and effective. The complete data including response after consolidation will be reported at the ASH meeting. This EMN trial was registered as NTR2422. Carfilzomib and an unrestricted grant was provided by ONYX Pharmaceuticals. Disclosures: Sonneveld: Millennium Pharmaceuticals, Inc.: Consultancy, Research Funding; Celgene: Consultancy, Research Funding; Janssen: Consultancy, Research Funding; Onyx: Consultancy, Research Funding. Zweegman:Celgene: Honoraria, Research Funding; Janssen-Cilag: Honoraria, Research Funding. Palumbo:Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Merck: Honoraria; Amgen: Honoraria.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 3470-3470
    Abstract: The HOVON-65/GMMG-HD4 trial is a prospective, randomized phase III trial to evaluate the efficacy of bortezomib prior to high-dose melphalan (200 mg/m2, HDM) on response and progression-free survival (PFS) in patients with newly diagnosed MM stage II or III according to Salmon & Durie (SD). Until May 2008 in total 833 patients aged 18–65 years were included in the trial. Patients were randomized to receive three cycles of VAD (arm A; vincristin 0,4mg, days 1–4, adriamycin 9 mg/m2, days 1–4, dexamethasone 40 mg, days 1–4, 9–12, and 17–20) or PAD (arm B; bortezomib 1.3 mg/m2, days 1,4,8,11, adriamycin 9 mg/m2, days 1–4, dexamethasone 40 mg, days 1–4, 9–12, and 17–20). Hematopoietic stem cells were mobilized in patients using the CAD regimen (cyclophosphamide 1000 mg/m2 iv day 1, adriamycin 15mg/m2, days 1–4, dexamethasone 40mg, days 1–4) and G-CSF. After stem cell harvesting all patients received one or two cycles of HDM with autologous stem cell transplantation followed by maintenance therapy with thalidomide 50 mg daily (arm A) and bortezomib 1.3 mg/m2 once every 2 weeks (arm B), respectively. As of August 2008 stem cell harvesting data from the first consecutively enrolled 150 patients (75 per arm) were analyzed. The data of the initial 300 patients (150 per arm) will be available by November 2008. Both treatment arms did not differ in age, SD stage of disease, ISS stage, and FISH abnormalities. 132 patients (88%) were treated with CAD plus G-CSF. Dosing and type of G-CSF treatment were comparable in both arms. In all patients stem cell collection was successful and at least two autografts could be harvested (minimal number of stem cells permitted per autograft was 2.0 ×106 CD 34+ cells per kg BW). Induction treatment Days until first leukapheresis Median (range) Number of leukaphereses Median (range) Number of harvested CD34+ stem cells (× 106) per kg BW Median (range) PAD 110 (96–149) 1 (1–4) 10.5 (4.1–37.6) VAD 111 (95–156) 1 (1–5) 9.3 (4.0–37.0) In 64% of the patients in arm A (VAD) and 79% of the patients in arm B (PAD) only one leukapheresis was sufficient for stem cell harvesting. In all patients hematopoietic reconstitution was achieved after HDM followed by autografts harvested after PAD or VAD. We conclude that stem cell harvesting after PAD is very well feasible.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 2132-2132
    Abstract: The value of Allo-SCT in myeloma is heavily disputed. In our previous Donor versus No Donor (DvND) comparison we found no survival benefit of Allo-RIC in newly diagnosed myeloma. (Lokhorst et al:Blood 2012119: 6219-6225). However, a recent update of the EBMT-NMA 2000 trial (Gahrton el al:Blood 2013121: 5055-506) suggested that extended follow-up ( 〉 5 years) may be necessary for a correct interpretation of a potential survival benefit for Allo-RIC. Here we present the extended follow-up of our trial, in which the median follow-up of patients now exceeds over 7.5 years since the first autologous SCT. Methods Patients with an HLA–identical sibling donor included in the phase III HOVON-50 study, that was designed to assess the role of thalidomide in induction treatment and maintenance after high-dose therapy (HDM 200 mg/m2), could proceed to the Hovon 54 study in which an Allo-SCT was performed after conditioning with low dose TBI only, between 2-6 months after HDM. Among the 536 eligible patients randomized in the HOVON-50 trial, ultimately 260 patients were eligible to be included into the DvND analysis: 122 patients with a donor, of whom 99 patients received an Allo-RIC and 138 without a donor, of whom 115 patients started maintenance therapy with thalidomide. Groups were comparable with regard to age, myeloma stage, and prognostic factors including cytogenetics and ISS stage. Results 93% of the patients in the no donor group achieved at least a PR (38% CR, 71% at least VGPR ), versus 96% of the patients in the donor group (43% CR, 73% at least VGPR). After a median follow-up of 91 months after HDM, PFS and OS were comparable between the two groups. In the intention to treat analysis median PFS was 29 months for the no donor group and 30 months the no donor group (P=0.25). Median OS was 76 for the donor group and 81 months for the no donor group (P=0.61). For the patients who actually received their allocated treatment (Allo-RIC or maintenance), PFS curves started to diverge after 3 years, however no statistical difference was observed (P=0.07). Allo-RIC improved the median overall survival from 73 to 94 months compared to patients receiving maintenance. However, due to frequent late mortality ( 〉 after 96 months) in the Allo-RIC group the benefit was not statistically significant (P=0.54). No subgroup including those achieving CR or those with high risk features (ISS, deletion of chromosome 13) did benefit from Allo-RIC. Conclusion This analysis failed to show improvement of tandem Auto Allo-RIC as part of first line therapy in myeloma as compared to Auto-SCT followed by maintenance therapy, even after extended follow-up. Disclosures: Lokhorst: Genmab A/S: Consultancy, Research Funding; Celgene: Honoraria; Johnson-Cilag: Honoraria; Mudipharma: Honoraria. Minnema:Janssen Cilag: Consultancy, Honoraria. Sonneveld:Janssen-Cilag: Honoraria; Celgene: Honoraria; Onyx: Honoraria; Janssen-Cilag: Research Funding; Millenium: Research Funding; Onyx: Research Funding; Celgene: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 1826-1826
    Abstract: Introduction: Daratumumab (DARA) has potent single agent activity in relapsed/refractory multiple myeloma (RRMM). The effectivity of DARA is partially dependent on the expression of its target, CD38, on tumor cells. Upregulation of CD38 on MM cells by all-trans retinoic acid (ATRA) improved the ex vivo efficacy of DARA, even in DARA-resistant MM cells. We therefore evaluated the efficacy and safety of DARA combined with ATRA in RRMM patients in the DARA/ATRA study (NCT02751255). Methods: In part A of this prospective, multicenter phase 1/2 trial, DARA was administered according to the approved schedule (16 mg/kg; cycles 1-2: weekly; cycles 3-6: biweekly; cycles ≥7: monthly). In part B, ATRA was given twice daily during 3 days prior to each DARA infusion. DARA dosing was resumed according to the aforementioned schedule. Inclusion criteria were ≥2 prior lines of treatment, WHO performance score 0-2, adequate bone marrow reserve, kidney and hepatic function. Patients were enrolled in part B, after treatment in part A, if they developed disease progression (PD), or if they had insufficient response defined as 〈 minimal response (MR) after cycle 2, or 〈 partial response (PR) after cycle 3, and if they still fulfilled the other inclusion criteria. All patients gave written informed consent. In phase 1 of part B, we determined the maximum tolerated dose (MTD) of ATRA (dose levels: 15, 30 and 45mg/m2) and the recommended dose level (RDL) for phase 2. We here report overall response rate (ORR), progression free survival (PFS(A)), overall survival (OS) and adverse events (AEs) for all patients in part A. For patients treated at the RDL in part B, we report ORR, PFS(A), PFS for part A + B combined (PFS(A+B)) and AEs. Results: Between July 2016 and October 2017, 63 patients were enrolled in part A, baseline characteristics are described in Table 1. At data cut-off (July 1st, 2019), 6 patients were still on treatment in part A. The median duration of follow-up of surviving patients at data cut-off was 26 months (range 18.2 - 39.4 months). The ORR in part A was 41% (22% PR, 14% very good PR [VGPR] and 5% (stringent) complete remission; Figure 1). The median PFS(A) and OS from start of DARA for all enrolled patients were 7.0 months (95% CI 5.3-8.8) and 28.2 months (95% CI 20.4-36.0), respectively. Eight patients did not meet eligibility criteria for part B, and 1 patient refused further treatment in part B. In part B phase 1, 14 patients were treated with ATRA at the 3 dose levels. The MTD was not reached and the RDL for phase 2 was defined as 45mg/m2. At data cut-off, 34 patients were enrolled in part B phase 2, of whom 2 patients were still on treatment. Thirty-two patients stopped treatment due to PD (n=28) or withdrawal of consent (n=4). Next, we focused on all 42 patients who were treated at the RDL in part B (8 patients in phase 1, and 34 patients in phase 2). The ORR in part B was 5% (VGPR n=2). However, 57% achieved MR or stable disease (SD; Figure 1). During part A, median PFS(A) in these patients was 6.7 months (95% CI 2.7-10.6). When stratified by ≥PR vs SD/MR vs PD/not evaluable (NE), median PFS(A) was 9.9 months (95% CI 3.1-16.7) vs 3.2 months (95% CI 2.5-3.9) vs 1.2 months (95% CI 0.8-1.5), respectively. Addition of ATRA and re-intensification of DARA in DARA-refractory patients resulted in a median PFS(A+B) of 7.9 months (95% CI 5.6-10.1 months). When stratified by ≥PR vs SD/MR vs PD/NE in part A, median PFS(A+B) was 17.7 months (95% CI 9.6-25.7) vs 5.4 months (95% CI 3.8-7.0) vs 2.5 months (95% CI 2.1-2.9), respectively. The presence of extramedullary plasmacytomas (P=0.004 and P=0.012) and WHO ≥ 1 (P=0.002 and P=0.005) were associated with lower PFS(A) and PFS(A+B). Importantly, combination of ATRA with DARA did not increase the rate of AEs (Table 2). The incidence of any grade infusion related reactions (IRR) was 34.9%. There were no complications with red blood cell transfusions. Conclusion: Here, we report for the first time on the efficacy of DARA re-intensification, combined with ATRA in DARA-refractory patients. Overall, this approach resulted in a modest prolongation of PFS. Patients with primary DARA-refractory disease did not benefit from the addition of ATRA. However, in patients with ≥PR on DARA monotherapy prior to progression, addition of ATRA and re-intensification of DARA was of marked clinical benefit with an additional 7.8 months of disease control. The combination of DARA and ATRA was well tolerated and safe. Disclosures Minnema: Celgene Corporation: Honoraria, Research Funding; Amgen: Honoraria; Gilead: Honoraria; Servier: Honoraria; Jansen Cilag: Honoraria. Levin:Takeda: Membership on an entity's Board of Directors or advisory committees, Other: Educational grant ; Amgen: Membership on an entity's Board of Directors or advisory committees, Other: Educational grant ; Janssen: Membership on an entity's Board of Directors or advisory committees, Other: Educational Grant; Roche: Membership on an entity's Board of Directors or advisory committees, Other: Educational Grant; Abbvie: Membership on an entity's Board of Directors or advisory committees, Other: Educational Grant. Broyl:Celgene, amgen, Janssen,Takeda: Honoraria. Bos:Celgene: Research Funding; Kiadis Pharma: Other: Shareholder (of Cytosen, acquired by Kiadis). Kersten:Miltenyi: Honoraria; Mundipharma: Honoraria, Research Funding; Kite Pharma: Honoraria, Research Funding; Bristol-Myers Squibb: Honoraria, Research Funding; Gilead: Honoraria; Amgen: Honoraria, Research Funding; Takeda Oncology: Research Funding; Celgene: Honoraria, Research Funding; Roche: Honoraria, Research Funding; Novartis: Honoraria. Mutis:Janssen Research and Development: Research Funding; Celgene: Research Funding; Onkimmune: Research Funding; Genmab: Research Funding. Zweegman:Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Research Funding. Sonneveld:SkylineDx: Research Funding; Takeda: Honoraria, Research Funding; Karyopharm: Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Celgene: Honoraria, Research Funding; BMS: Honoraria; Amgen: Honoraria, Research Funding. Van De Donk:Bristol-Myers Squibb: Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Membership on an entity's Board of Directors or advisory committees; Janssen Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Servier: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees; Bayer: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; AMGEN: Membership on an entity's Board of Directors or advisory committees, Research Funding. OffLabel Disclosure: ATRA for treatment of MM
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages