19 research outputs found

    A Comprehensive Patient-Derived Xenograft Collection Representing the Heterogeneity of Melanoma

    Get PDF
    Therapy of advanced melanoma is changing dramatically. Following mutational and biological subclassification of this heterogeneous cancer, several targeted and immune therapies were approved and increased survival significantly. To facilitate further advancements through pre-clinical in vivo modeling, we have established 459 patient-derived xenografts (PDX) and live tissue samples from 384 patients representing the full spectrum of clinical, therapeutic, mutational, and biological heterogeneity of melanoma. PDX have been characterized using targeted sequencing and protein arrays and are clinically annotated. This exhaustive live tissue resource includes PDX from 57 samples resistant to targeted therapy, 61 samples from responders and non-responders to immune checkpoint blockade, and 31 samples from brain metastasis. Uveal, mucosal, and acral subtypes are represented as well. We show examples of pre-clinical trials that highlight how the PDX collection can be used to develop and optimize precision therapies, biomarkers of response, and the targeting of rare genetic subgroups

    The Public Repository of Xenografts enables discovery and randomized phase II-like trials in mice

    Get PDF
    More than 90% of drugs with preclinical activity fail in human trials, largely due to insufficient efficacy. We hypothesized that adequately powered trials of patient-derived xenografts (PDX) in mice could efficiently define therapeutic activity across heterogeneous tumors. To address this hypothesis, we established a large, publicly available repository of well-characterized leukemia and lymphoma PDXs that undergo orthotopic engraftment, called the Public Repository of Xenografts (PRoXe). PRoXe includes all de-identified information relevant to the primary specimens and the PDXs derived from them. Using this repository, we demonstrate that large studies of acute leukemia PDXs that mimic human randomized clinical trials can characterize drug efficacy and generate transcriptional, functional, and proteomic biomarkers in both treatment-naive and relapsed/refractory disease

    Pharmacokinetic–pharmacodynamic guided optimisation of dose and schedule of CGM097, an HDM2 inhibitor, in preclinical and clinical studies

    Full text link
    Background: CGM097 inhibits the p53-HDM2 interaction leading to downstream p53 activation. Preclinical in vivo studies support clinical exploration while providing preliminary evidence for dosing regimens. This first-in-human phase I study aimed at assessing the safety, MTD, PK/PD and preliminary antitumor activity of CGM097 in advanced solid tumour patients (NCT01760525). Methods: Fifty-one patients received oral treatment with CGM097 10-400 mg 3qw (n = 31) or 300-700 mg 3qw 2 weeks on/1 week off (n = 20). Choice of dose regimen was guided by PD biomarkers, and quantitative models describing the effect of CGM097 on circulating platelet and PD kinetics. Results: No dose-limiting toxicities were reported in any regimens. The most common treatment-related grade 3/4 AEs were haematologic events. PK/PD models well described the time course of platelet and serum GDF-15 changes, providing a tool to predict response to CGM097 for dose-limiting thrombocytopenia and GDF-15 biomarker. The disease control rate was 39%, including one partial response and 19 patients in stable disease. Twenty patients had a cumulative treatment duration of >16 weeks, with eight patients on treatment for >32 weeks. The MTD was not determined. Conclusions: Despite delayed-onset thrombocytopenia frequently observed, the tolerability of CGM097 appears manageable. This study provided insights on dosing optimisation for next-generation HDM2 inhibitors. Translational relevance: Haematologic toxicity with delayed thrombocytopenia is a well-known on-target effect of HDM2 inhibitors. Here we have developed a PK/PD guided approach to optimise the dose and schedule of CGM097, a novel HDM2 inhibitor, using exposure, platelets and GDF-15, a known p53 downstream target to predict patients at higher risk to develop thrombocytopenia. While CGM097 had shown limited activity, with disease control rate of 39% and only one patient in partial response, the preliminary data from the first-in-human escalation study together with the PK/PD modeling provide important insights on how to optimize dosing of next generation HDM2 inhibitors to mitigate hematologic toxicity

    High-mobility organic thin-film transistors based on a small-molecule semiconductor deposited in vacuum and by solution shearing

    No full text
    The small-molecule organic semiconductor 2,9-di-decyl-dinaphtho-[2,3-b: 2',3'-f]-thieno[3,2-b]-thiophene (C-10-DNTT) was used to fabricate bottom-gate, top-contact thin-film transistors (TFTs) in which the semiconductor layer was prepared either by vacuum deposition or by solution shearing. The maximum effective charge-carrier mobility of TFTs with vacuum-deposited C-10-DNTT is 8.5 cm(2)/V s for a nominal semiconductor thickness of 10 nm and a substrate temperature during the semiconductor deposition of 80 degrees C. Scanning electron microscopy analysis reveals the growth of small, isolated islands that begin to coalesce into a flat conducting layer when the nominal thickness exceeds 4 nm. The morphology of the vacuum-deposited semiconductor layers is dominated by tall lamellae that are formed during the deposition, except at very high substrate temperatures. Atomic force microscopy and X-ray diffraction measurements indicate that the C-10-DNTT molecules stand approximately upright with respect to the substrate surface, both in the flat conducting layer near the surface and within the lamellae. Using the transmission line method on TFTs with channel lengths ranging from 10 to 100 mu m, a relatively small contact resistance of 0.33 k Omega cm was determined. TFTs with the C-10-DNTT layer prepared by solution shearing exhibit a pronounced anisotropy of the electrical performance: TFTs with the channel oriented parallel to the shearing direction have an average carrier mobility of (2.8 +/- 0.3) cm(2)/V s, while TFTs with the channel oriented perpendicular to the shearing direction have a somewhat smaller average mobility of (1.3 +/- 0.1) cm(2)/V s. (C) 2013 Elsevier B.V. All rights reserved

    The HDM2 (MDM2) Inhibitor NVP-CGM097 inhibits tumor cell proliferation and shows additive effects with 5-fluorouracil on the p53 - p21 - Rb - E2F1 cascade in the p53wildtype neuroendocrine tumor cell line GOT1

    No full text
    Background/aims: The tumor suppressor p53 is depleted in many tumor cells by the E3 ubiquitin ligase mouse double minute 2 homolog (MDM2) through MDM2/p53 interaction. A novel target for inhibiting p53 degradation and for causing reexpression of p53wild type is inhibition of MDM2. The small molecule NVP-CGM097 is a novel MDM2 inhibitor. We investigated MDM2 inhibition as a target in neuroendocrine tumor cells in vitro. Methods: Human neuroendocrine tumor cell lines from the pancreas (BON1), lung (NCI-H727), and midgut (GOT1) were incubated with the MDM2 inhibitor NVP-CGM097 (Novartis) at concentrations from 4 to 2,500 nM. Results: While p53wild type GOT1 cells were sensitive to NVP-CGM097, p53mutated BON1 and p53mutated NCI-H727 cells were resistant to NVP-CGM097. Incubation of GOT1 cells with NVP-CGM097 at 100, 500, and 2,500 nM for 96 h caused a significant decline in cell viability to 84.9 ± 9.2% (p < 0.05), 77.4 ± 6.6% (p < 0.01), and 47.7 ± 9.2% (p < 0.01). In a Western blot analysis of GOT1 cells, NVP-CGM097 caused a dose-dependent increase in the expression of p53 and p21 tumor suppressor proteins and a decrease in phospho-Rb and E2F1. Experiments of co-incubation of NVP-CGM097 with 5-fluorouracil, temozolomide, or everolimus each showed additive antiproliferative effects in GOT1 cells. NVP-CGM097 and 5-fluorouracil increased p53 and p21 expression in an additive manner. Conclusions: MDM2 inhibition seems a promising novel therapeutic target in neuroendocrine tumors harboring p53wild type. Further investigations should examine the potential role of MDM2 inhibitors in neuroendocrine tumor treatment. Keywords: 5-Fluorouracil; Everolimus; GOT1; MDM2 inhibitor; NVP-CGM097; Neuroendocrine tumor; Temozolomide; p21; p53

    First-in-Human, Phase I Dose-Escalation Study of CGM097, a HDM2 Inhibitor in Adult Patients With p53 Wild-type Advanced Solid Malignancies

    No full text
    Background: CGM097 inhibits p53-HDM2 interaction, thus activates p53 downstream effector pathways inducing cell cycle arrest and/or apoptosis. This phase I study aimed at assessing the safety, MTD, PK/PD, and preliminary antitumor activity of CGM097 in advanced solid tumors patients (NCT01760525). Methods: Fifty-one patients received oral treatment with CGM097 10-400mg 3qw (n=31) and on an alternative regimen of 3qw 2 weeks on/1 week off (300-700mg; n=20). Choice of dose regimen was guided by PD biomarkers, PK/PD relationship, and modeling of drug-induced changes in platelet kinetics. Results: No dose-limiting toxicities were reported in any regimens. Grade 3/4 AEs suspected to be drug-related were reported in 21 patients. Main reason for discontinuation was disease progression (n=41). CGM097 plasma concentrations increased in a dose proportional manner. Disease control rate was 39%, including one partial response (300mg, 3qw) and 19 patients in stable disease (10-700mg 3qw). Forty patients had a cumulative treatment duration of >16 weeks, with 8 patients on treatment for >32 weeks. The MTD was not determined. Conclusions: Despite delayed-onset thrombocytopenia frequently observed, the tolerability of CGM097 appears manageable. Although CGM097 is not being further developed, this study led to important modeling-derived learnings to optimize dose scheduling of next generation HDM2 inhibitors

    Inhibition of wild-type p53-expressing AML by novel small molecule HDM2 inhibitor, CGM097

    No full text
    The tumor suppressor, p53, is a key regulator of apoptosis and functions upstream in the apoptotic cascade by both indirectly and directly regulating Bcl-2 family proteins. In cells expressing wild-type (wt) p53, the Human Double Minute 2 (HDM2) protein binds to p53 and blocks its activity. Inhibition of HDM2:p53 interaction activates p53 and causes apoptosis or cell cycle arrest. We have characterized the novel HDM2 inhibitor, CGM097, as having significant activity against wt p53-expressing acute myeloid leukemia (AML). Specifically, CGM097 potently and selectively inhibited the proliferation of human AML cell lines and primary AML cells expressing wt p53, but not mutant p53, in a target-specific manner. Several patient samples that harbored mutant p53 were comparatively unresponsive to CGM097. Synergy was observed when CGM097 was combined with FLT3 inhibition against mutant FLT3-expressing cells, as well as when combined with MEK inhibition in cells with activated MAPK signaling. Finally, CGM097 was effective in reducing leukemia burden in vivo. Taken together, these data suggest that CGM097 might be a promising treatment for AML characterized as harboring wt p53 as a single agent, as well as possibly in combination with another targeted therapy using tyrosine kinase inhibitors (TKIs) against oncogenes that drive AML

    Pharmacokinetic–pharmacodynamic guided optimisation of dose and schedule of CGM097, an HDM2 inhibitor, in preclinical and clinical studies

    No full text
    Background!#!CGM097 inhibits the p53-HDM2 interaction leading to downstream p53 activation. Preclinical in vivo studies support clinical exploration while providing preliminary evidence for dosing regimens. This first-in-human phase I study aimed at assessing the safety, MTD, PK/PD and preliminary antitumor activity of CGM097 in advanced solid tumour patients (NCT01760525).!##!Methods!#!Fifty-one patients received oral treatment with CGM097 10-400 mg 3qw (n = 31) or 300-700 mg 3qw 2 weeks on/1 week off (n = 20). Choice of dose regimen was guided by PD biomarkers, and quantitative models describing the effect of CGM097 on circulating platelet and PD kinetics.!##!Results!#!No dose-limiting toxicities were reported in any regimens. The most common treatment-related grade 3/4 AEs were haematologic events. PK/PD models well described the time course of platelet and serum GDF-15 changes, providing a tool to predict response to CGM097 for dose-limiting thrombocytopenia and GDF-15 biomarker. The disease control rate was 39%, including one partial response and 19 patients in stable disease. Twenty patients had a cumulative treatment duration of &amp;gt;16 weeks, with eight patients on treatment for &amp;gt;32 weeks. The MTD was not determined.!##!Conclusions!#!Despite delayed-onset thrombocytopenia frequently observed, the tolerability of CGM097 appears manageable. This study provided insights on dosing optimisation for next-generation HDM2 inhibitors.!##!Translational relevance!#!Haematologic toxicity with delayed thrombocytopenia is a well-known on-target effect of HDM2 inhibitors. Here we have developed a PK/PD guided approach to optimise the dose and schedule of CGM097, a novel HDM2 inhibitor, using exposure, platelets and GDF-15, a known p53 downstream target to predict patients at higher risk to develop thrombocytopenia. While CGM097 had shown limited activity, with disease control rate of 39% and only one patient in partial response, the preliminary data from the first-in-human escalation study together with the PK/PD modeling provide important insights on how to optimize dosing of next generation HDM2 inhibitors to mitigate hematologic toxicity

    Inhibition of wild-type p53-expressing AML by the novel small molecule HDM2 inhibitor CGM097

    No full text
    The tumor suppressor p53 is a key regulator of apoptosis and functions upstream in the apoptotic cascade by both indirectly and directly regulating Bcl-2 family proteins. In cells expressing wild-type (WT) p53, the HDM2 protein binds to p53 and blocks its activity. Inhibition of HDM2:p53 interaction activates p53 and causes apoptosis or cell-cycle arrest. Here, we investigated the ability of the novel HDM2 inhibitor CGM097 to potently and selectively kill WT p53-expressing AML cells. The antileukemic effects of CGM097 were studied using cellbased proliferation assays (human AML cell lines, primary AML patient cells, and normal bone marrow samples), apoptosis, and cell-cycle assays, ELISA, immunoblotting, and an AML patient-derived in vivo mouse model. CGM097 potently and selectively inhibited the proliferation of human AML cell lines and the majority of primary AML cells expressing WT p53, but not mutant p53, in a target-specific manner. Several patient samples that harbored mutant p53 were comparatively unresponsive to CGM097. Synergy was observed when CGM097 was combined with FLT3 inhibition against oncogenic FLT3-expressing cells cultured both in the absence as well as the presence of cytoprotective stromal-secreted cytokines, as well as when combined with MEK inhibition in cells with activated MAPK signaling. Finally, CGM097 was effective in reducing leukemia burden in vivo. These data suggest that CGM097 is a promising treatment for AML characterized as harboring WT p53 as a single agent, as well as in combination with other therapies targeting oncogene-activated pathways that drive AML
    corecore