17 research outputs found

    A phase 1b study of venetoclax and alvocidib in patients with relapsed/refractory acute myeloid leukemia

    Get PDF
    Relapsed/refractory (R/R) Acute Myeloid Leukemia (AML) is a genetically complex and heterogeneous disease with a poor prognosis and limited treatment options. Thus, there is an urgent need to develop therapeutic combinations to overcome drug resistance in AML. This open-label, multicenter, international, phase 1b study evaluated the safety, efficacy, and pharmacokinetics of venetoclax in combination with alvocidib in patients with R/R AML. Patients were treated with escalating doses of venetoclax (400, 600, and 800 mg QD, orally, days 1–28) and alvocidib (45 and 60 mg/m2, intravenously, days 1–3) in 28-day cycles. The combination was found to be safe and tolerable, with no maximum tolerated dose reached. Drug-related Grade ≥3 adverse events were reported in 23 (65.7%) for venetoclax and 24 (68.6%) for alvocidib. No drug-related AEs were fatal. Gastrointestinal toxicities, including diarrhea, nausea, and vomiting were notable and frequent; otherwise, the toxicities reported were consistent with the safety profile of both agents. The response rate was modest (complete remission [CR] + incomplete CR [CRi], 11.4%; CR + CRi + partial response rate + morphologic leukemia-free state, 20%). There was no change in alvocidib pharmacokinetics with increasing doses of venetoclax. However, when venetoclax was administered with alvocidib, AUC24 and Cmax decreased by 18% and 19%, respectively. A recommended phase 2 dose was not established due to lack of meaningful increase in efficacy across all cohorts compared to what was previously observed with each agent alone. Future studies could consider the role of the sequence, dosing, and the use of a more selective MCL1 inhibitor for the R/R AML population

    Discovery of Genetic Mechanisms Underlying Inter-Individual Differences in Allopurinol Response

    No full text
    Reverse translational research takes a bedside to bench approach, using sophisticated basic research to explain the biological mechanisms behind observed clinical data. Here, we used this approach to characterize the inter-individual differences in the efficacy and pharmacokinetics of a commonly prescribed drug, allopurinol. Allopurinol is the first-line treatment for chronic gout, a debilitating arthritis caused by high serum uric acid levels. Not only is the prevalence of gout growing rapidly, but high serum uric acid and gout have been shown to be important risk factors for the development of comorbidities such as renal and cardiovascular disease. Thus, allopurinol is increasingly being used in preventing these deadly diseases. However, response to allopurinol is variable, and many gout patients fail to achieve healthy serum uric acid levels, even while adherent. Our reverse translational approach begins with the largest genome-wide association study on allopurinol response to date, performed in a multi-ethnic cohort of patients taking allopurinol for the treatment of gout. Consistent with previous studies, we observed that the Q141K variant in ABCG2 (rs2231142), which encodes the efflux pump BCRP, associated with worse response to allopurinol. However, for the first time this association reached genome-wide level significance (p=8.06 x 10-11). Additionally, we identified a novel association with a variant in GREM2 (rs1934341, p=3.2 x 10-6). In vitro studies led to the identification of oxypurinol, the active metabolite of allopurinol, as an inhibitor of the uric acid transporter GLUT9, suggesting that oxypurinol may modulate uric acid disposition. These results suggest that allopurinol may have additional hypouricemic effects beyond xanthine oxidase inhibition, a mechanism previously unknown. In order to further explore the role between BCRP Q141K and allopurinol response, we performed a clinical study in which we characterized the relationship between BCRP Q141K and allopurinol pharmacokinetics and pharmacodynamics. We observed a significant difference in oxypurinol half-life in subjects harboring the Q141K variant (19.1 ± 0.5 vs 23.3 ± 0.2, p=0.02), but no differences in any other pharmacokinetic parameters. These results suggest that the association between response and BCRP Q141K may be dependent on tissue levels of the drug, rather than systemic levels. These findings have important implications in the understanding of allopurinol’s mechanism of action and clinical use, and more broadly, for the complex relationships between BCRP Q141K and the pharmacokinetics of its substrates

    Oxypurinol pharmacokinetics and pharmacodynamics in healthy volunteers: Influence of BCRP Q141K polymorphism and patient characteristics.

    No full text
    The missense variant, breast cancer resistance protein (BCRP) p.Q141K, which encodes a reduced function BCRP, has been linked to poor response to allopurinol. Using a multifaceted approach, we aimed to characterize the relationship(s) between BCRP p.Q141K, the pharmacokinetics (PK) and pharmacodynamics (PD) of oxypurinol (the active metabolite of allopurinol), and serum uric acid (SUA) levels. A prospective clinical study (NCT02956278) was conducted in which healthy volunteers were given a single oral dose of 300 mg allopurinol followed by intensive blood sampling. Data were analyzed using noncompartmental analysis and population PK/PD modeling. Additionally, electronic health records were analyzed to investigate whether clinical inhibitors of BCRP phenocopied the effects of the p.Q141K variant with respect to SUA. Subjects homozygous for p.Q141K had a longer half-life (34.2 ± 12.2 h vs. 19.1 ± 1.42 h) of oxypurinol. The PK/PD model showed that women had a 24.8% lower volume of distribution. Baseline SUA was affected by p.Q141K genotype and renal function; that is, it changed by 48.8% for every 1 mg/dl difference in serum creatinine. Real-world data analyses showed that patients prescribed clinical inhibitors of BCRP have higher SUA levels than those that have not been prescribed inhibitors of BCRP, consistent with the idea that BCRP inhibitors phenocopy the effects of p.Q141K on uric acid levels. This study identified important covariates of oxypurinol PK/PD that could affect its efficacy for the treatment of gout as well as a potential side effect of BCRP inhibitors on increasing uric acid levels, which has not been described previously

    A conserved role of the insulin-like signaling pathway in diet-dependent uric acid pathologies in Drosophila melanogaster.

    No full text
    Elevated uric acid (UA) is a key risk factor for many disorders, including metabolic syndrome, gout and kidney stones. Despite frequent occurrence of these disorders, the genetic pathways influencing UA metabolism and the association with disease remain poorly understood. In humans, elevated UA levels resulted from the loss of the of the urate oxidase (Uro) gene around 15 million years ago. Therefore, we established a Drosophila melanogaster model with reduced expression of the orthologous Uro gene to study the pathogenesis arising from elevated UA. Reduced Uro expression in Drosophila resulted in elevated UA levels, accumulation of concretions in the excretory system, and shortening of lifespan when reared on diets containing high levels of yeast extract. Furthermore, high levels of dietary purines, but not protein or sugar, were sufficient to produce the same effects of shortened lifespan and concretion formation in the Drosophila model. The insulin-like signaling (ILS) pathway has been shown to respond to changes in nutrient status in several species. We observed that genetic suppression of ILS genes reduced both UA levels and concretion load in flies fed high levels of yeast extract. Further support for the role of the ILS pathway in modulating UA metabolism stems from a human candidate gene study identifying SNPs in the ILS genes AKT2 and FOXO3 being associated with serum UA levels or gout. Additionally, inhibition of the NADPH oxidase (NOX) gene rescued the reduced lifespan and concretion phenotypes in Uro knockdown flies. Thus, components of the ILS pathway and the downstream protein NOX represent potential therapeutic targets for treating UA associated pathologies, including gout and kidney stones, as well as extending human healthspan
    corecore