Bortezomib, vorinostat and dexamethasone combination therapy in relapsed myeloma: results of the phase 2 MUK trial WQZa

Author(s):  
Sarah R. Brown ◽  
Charlotte Pawlyn ◽  
Avie-Lee Tillotson ◽  
Debbie Sherratt ◽  
Louise Flanagan ◽  
...  
2017 ◽  
Vol 4 (suppl_1) ◽  
pp. S366-S367 ◽  
Author(s):  
Ian Morrissey ◽  
Sophie Magnet ◽  
Stephen Hawser ◽  
Stuart Shapiro ◽  
Harald Seifert ◽  
...  

Abstract Background AAI101 is a novel extended-spectrum β-lactamase inhibitor (BLI), active against ESBLs and a broad array of other BLs. AAI101 in combination with cefepime (FEP) is in Phase 2 development. Infections caused by A. baumannii, a pathogen endemic to the southern US and other global regions, are very challenging to treat, and often require combination therapy. This study examined the activity of FEP/AAI101 against a challenge set of A. baumannii clinical isolates enriched with OXA carbapenemase producers. Methods BLs in A. baumannii were identified by genotyping. Broth microdilution MICs and susceptibilities were obtained following CLSI methods and breakpoints (BPs), except for ceftazidime-avibactam (CAZ/AVI) where FDA P. aeruginosa BPs were used. CLSI FEP BPs were used for FEP/AAI101. Results All OXA-51 producers had the ISAba1 promoter. MIC90 data and % susceptibilities (%S) for FEP/AAI101 and comparators are shown in the Table: FEP/AAI101 was highly active against meropenem-susceptible (MPMs) isolates. FEP/AAI101 (AAI101 fixed at 8 µg/ml) covered 67% of OXA-51 and 53% of OXA-58 strains. Lower susceptibilities were obtained for OXA-23 and OXA-24/40 producers. FEP/AAI101 was the most active β-lactam product. Colistin (COL) was the only agent with consistently high activity against all A. baumannii isolates. Conclusion FEP/AAI101 was the most potent β-lactam product tested against clinical isolates of A. baumannii producing OXA-51 and OXA-58 β-lactamases. Infections by this difficult pathogen often require combination therapy, of which FEP-AAI101 may be a component. Disclosures S. Shapiro, Allecra: Employee, Salary


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1852-1852
Author(s):  
Matthew W Jenner ◽  
Avie-Lee Tillotson ◽  
Sarah R Brown ◽  
Louise M Flanagan ◽  
Debbie Sherratt ◽  
...  

Abstract Introduction: Bortezomib (Velcade) and dexamethasone is a standard combination for relapsed myeloma. Both in vitro data and initial clinical trials signalled the efficacy of the combination of intravenous bortezomib and the oral histone deacetylase inhibitor vorinostat. Although the randomised phase 3 VANTAGE 088 trial identified an improvement in progression free survival with the combination of bortezomib and vorinostat compared to bortezomib monotherapy in relapsed myeloma, 50% of patients in the vorinostat group had at least one dose reduction compared with 25% in the placebo group, with potential impact on clinical outcomes. Subcutaneous bortezomib has now become the standard route of administration because of lower rates of peripheral neuropathy. MUK four is a single arm phase 2 multi-centre UK trial to evaluate the toxicity profile and efficacy of an alternative dosing schedule of vorinostat in combination with subcutaneous bortezomib and oral dexamethasone. We report the final analysis of toxicity and response data. Methods: Patients with relapsed myeloma treated with 1-3 prior lines of therapy received up to 8 cycles of V2 D (bortezomib 1.3 mg/m2 subcutaneously days 1, 4, 8 and 11, vorinostat 400 mg orally days 1-4, 8-11 and 15-18 and dexamethasone 20 mg orally days 1, 2, 4, 5, 8, 9, 11 and 12 of a 21 day cycle). Following completion of a minimum of 3 cycles of V2 D, participants received maintenance vorinostat (400 mg days 1-4 and 15-18 of a 28 day cycle) until disease progression, intolerance or participant withdrawal. Responses were assessed using the modified IMWG response criteria and toxicities graded using CTCAE v4.0. Results: Between August 2013 and November 2014, 16 participants were recruited to MUK four. Median age was 69.5 years (range 50.0-78.0) and median lines of prior treatment was 1 (1-3). Prior treatment included thalidomide-based combinations in 13/16 (81.3%), bortezomib-based in 7/16 (43.8%) and lenalidomide-based in 2/16 (12.5%). 9/16 (56.3%) participants had received prior high dose melphalan ASCT. Median time from diagnosis was 38.6 months (9.3-120.4). At analysis in June 2015 8/16 (50%) participants continued on maintenance vorinostat. All 16 patients were evaluable for response within the first 8 cycles of V2 D. Overall response rate was 81.3% (13/16, 95% CI [55.4-96.0]) consisting of CR in 4/16 (25.0%), VGPR 2/16 (12.5%) and PR in 7/16 (43.8%). The remaining 3/16 (18.8%) achieved MR giving a clinical benefit response rate of 16/16 (100%). Participants received a median of 6 cycles of initial treatment with 6/16 (37.5%) receiving all 8 cycles. Treatment was discontinued in 4/8 (50%) because of disease progression, in 2/8 (25%) because of toxicity and in 2/8 (25%) for clinician discretion. Overall 12/16 (75%) participants experienced a dose reduction of either vorinostat or bortezomib or terminated treatment early as a result of toxicity. 11/16 (68.8%) reduced vorinostat and 10/16 (62.5%) reduced bortezomib. The most frequent grade 2 toxicities during the first 8 cycles were fatigue in 8/16 (50%), anaemia in 7/16 (43.8%), diarrhoea in 5/16 (31.3%), nausea in 4/16 (25.0%) and peripheral neuropathy in 4/16 (25.0%). The most frequent grade 3-4 toxicities encountered during the first 8 cycles were thrombocytopenia in 8/16 (50%), anaemia in 1/16 (6.3%), diarrhoea in 1/16 (6.3%) and fatigue in 1/16 (6.3%). During maintenance vorinostat only 1 participant experienced an adverse reaction above grade 2 (grade 3 neutropenia). Conclusion: Bortezomib, vorinostat and dexamethasone is a highly effective combination in relapsed myeloma with good response rates. Maintenance vorinostat is well tolerated. Although toxicity and dose reductions are observed with combination therapy, this study demonstrates that the combination of proteasome inhibitor, HDAC inhibitor and dexamethasone offers promise. Further data on PFS will be presented. Disclosures Jenner: Amgen: Honoraria; Takeda: Honoraria. Off Label Use: Vorinostat for treatment of myeloma. Pawlyn:Celgene: Honoraria, Other: Travel support; The Institute of Cancer Research: Employment. Williams:Celgene: Consultancy, Speakers Bureau; Janssen: Consultancy, Honoraria, Speakers Bureau; Amgen: Consultancy, Speakers Bureau; Takeda: Consultancy, Speakers Bureau. Davies:Array-Biopharma: Membership on an entity's Board of Directors or advisory committees; Takeda-Millennium: Membership on an entity's Board of Directors or advisory committees; University of Arkansas for Medical Sciences: Employment; Onyx-Amgen: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees.


2006 ◽  
Vol 14 (7S_Part_31) ◽  
pp. P1622-P1623 ◽  
Author(s):  
Michael C. Irizarry ◽  
Adam S. Fleisher ◽  
Ann Marie Hake ◽  
Peng Liu ◽  
Sergey Shcherbinin ◽  
...  

2017 ◽  
Vol 60 ◽  
pp. 31-35 ◽  
Author(s):  
K. Gowin ◽  
H. Kosiorek ◽  
A. Dueck ◽  
J. Mascarenhas ◽  
R. Hoffman ◽  
...  

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. TPS3141-TPS3141
Author(s):  
Ryo Kudo ◽  
Toshio Kubo ◽  
Yukiko Mori ◽  
Yohei Harada ◽  
Hidekazu Shirota ◽  
...  

TPS3141 Background: The human epidermal growth factor receptor 2 (HER2) gene amplification and mutations have emerged as oncogenic drivers and therapeutic targets not limited to breast and gastric cancers, but also in a variety of cancers. However, even if an actionable gene alteration is found, the incidence of HER2 amplification in these cancers is less than 5%. Despite its considerable therapeutic potential, the evidence is not yet mature enough for use as treatment in clinical practice. To address this unmet clinical need, we have designed an organ-agnostic basket trial, which covers a variety of solid cancers harboring HER2 amplification. Methods: JUPITER trial is a Japanese multicenter, single-arm, phase 2 basket study of combination therapy with trastuzumab and pertuzumab. Patients with solid cancers harboring HER2 amplification, who have progressed with standard treatment, or rare cancers for which there is no standard treatment, are eligible. Types of cancer include bile duct, urothelial, uterine, ovarian, and other solid cancers that HER2 amplification is detected by comprehensive genomic profiling using next-generation sequencing technology. Target sample size is 38. All enrolled patients receive combination therapy with trastuzumab and pertuzumab every 3 weeks until disease progression, unacceptable toxicity, death, or withdrawal of informed consent. Response assessment using RECIST version 1.1 is performed at weeks 9 and 17, followed by every 12 weeks. The primary endpoint is the objective response rate, and secondary endpoints are progression-free survival, overall survival, duration of response, and safety. In total, 40 patients were enrolled by June 2020. Data fix is scheduled in September 2021. Trial registration: jRCT2031180150 Clinical trial information: jRCT2031180150.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 26-27
Author(s):  
Qingqing Cai ◽  
Huiqiang Huang ◽  
Panpan Liu ◽  
Hui Zhou ◽  
Yajun Li ◽  
...  

Background: Extranodal natural killer/T cell lymphoma, nasal type (ENKTL) is a highly malignant non-Hodgkin lymphoma (NHL), which has no standard treatment available for this subgroup. Conventional treatment approaches (L-asparaginase- or pegaspargase-based regimen) failed to achieve long-term clinical benefit. The prognosis of advanced ENKTL is poor even in newly diagnosed patients (pts) due to a high relapse rate of 70%. Substantial evidences indicate that anti-programmed death-1 (PD-1) monoclonal antibody has achieved promising efficacy in refractory or relapsed ENKTL (Kwong et al. Blood 2017, Li et al. Journal of Hematology & Oncology 2018, Tao et al. JCO 2020). Results from our retrospective study demonstrated a manageable tolerability profile and favorable anti-tumor activity of anti-PD-1 antibody combined with P-GEMOX (pegaspargase, gemcitabine and oxaliplatin) regimen in untreated advanced ENKTL pts (unpublished data). Therefore, a phase 2 trial (NCT04127227) was initiated to evaluate safety and efficacy of sintilimab (a fully human anti-PD-1 monoclonal antibody) plus P-GEMOX regimen as first-line therapy in advanced ENKTL pts. Here, we report the preliminary results of the study. Methods: This study enrolled adult pts with untreated pathologically confirmed ENKTL with at least one measurable lesion. A sample size of 34 pts was preplanned, with 6 pts recruited for a safety run-in cohort. Pts received six 21-day cycles of sintilimab 200 mg, pegaspargase 2000 U/m2 (day 1), gemcitabine 1g/m2 (days 1 and 8), and oxaliplatin 130 mg/m2 (day 1), followed by sintilimab 200 mg maintenance every 3 weeks for up to 2 years or until documented confirmed disease progression or intolerable toxicity. Of the run-in cohort, preliminary safety and dose-limiting toxicity (DLTs) analyses were performed in the first cycle of treatment, primary endpoints were safety and tolerability of the combination therapy. DLTs were defined as grade 4 neutropenia that does not resolve to grade </= 2 within 7 days; grade 4 thrombocytopenia that does not resolve within 7 days; any grade >/= 3 nonhematologic toxicity lasting for more than 7 days, and any treatment-related adverse events (TRAEs) that results in a delay for more than 14 days of initiating cycle 2. Of the entire cohort, primary endpoints were complete response (CR) rate and objective response rate (ORR), secondary endpoints included overall survival, progression-free survival, and safety. TRAEs were accessed according to CTCAE 5.0. Response assessments were performed using PET/CT or MRI scans every 6 weeks of the combined treatment and every 3 months of maintenance treatment. Results: Six pts with untreated ENKTL were enrolled between Sept 19, 2019 and Jul 26, 2020. Median age was 56 years (range 52-67). All pts were found to be found to be positive for EBV encoded RNA (EBER) by in situ hybridization. Clinical characteristics of all pts are listed in Table 1. Median number of treatment cycles was 7 (range 4-11). At data cut-off, 5 pts completed 6 cycles of combination therapy and entered the maintenance phase (median: 1, range 1-5). No DLTs were observed. The majority of TRAEs were reported during the first two cycles of the combination treatment. The most common TRAEs were anorexia (6/6, 100%), elevated transaminase (6/6, 100%), neutropenia (5/6, 3.3%), nausea (4/6, 66.7%), anemia (4/6, 66.7%), hypoproteinemia (4/6, 66.7%), vomiting (3/6, 50.0%) and thrombocytopenia (3/6, 33.3%). Grade 3/4 TRAEs occurred in 4 (66.7%) pts, including thrombocytopenia (1/6, 16.7%), anemia (2/6, 33.0%), and neutropenia (1/6, 16.7%). Details of all-grade TRAEs are listed in Table 2. All the TRAEs were manageable and reversible. No immune-related (ir) AE or treatment-related death was observed. Of the 6 pts received response assessments, ORR was 100% (6/6) and CR rate was 33.3% (2/6). After a median follow-up of 7.8 months, 5 pts were still on the planned treatment and maintained favorable outcomes. The updated data will be reported at the ASH conference. Conclusions: Sintilimab plus P-GEMOX regimen demonstrated manageable safety profile and promising anti-tumor activity in pts with untreated advanced ENKTL. The preliminary safety and efficacy profile of combination therapy support further study and the exploration of effective biomarkers for predicting the treatment response is under way. Disclosures No relevant conflicts of interest to declare. OffLabel Disclosure: Sintilimab is medication that is indicated for the treatment of relapsed or refractory classical Hodgkin's lymphoma after failure of at least second-line systemic chemotherapy.


Sign in / Sign up

Export Citation Format

Share Document