17 research outputs found

    31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016) : part two

    Get PDF
    Background The immunological escape of tumors represents one of the main ob- stacles to the treatment of malignancies. The blockade of PD-1 or CTLA-4 receptors represented a milestone in the history of immunotherapy. However, immune checkpoint inhibitors seem to be effective in specific cohorts of patients. It has been proposed that their efficacy relies on the presence of an immunological response. Thus, we hypothesized that disruption of the PD-L1/PD-1 axis would synergize with our oncolytic vaccine platform PeptiCRAd. Methods We used murine B16OVA in vivo tumor models and flow cytometry analysis to investigate the immunological background. Results First, we found that high-burden B16OVA tumors were refractory to combination immunotherapy. However, with a more aggressive schedule, tumors with a lower burden were more susceptible to the combination of PeptiCRAd and PD-L1 blockade. The therapy signifi- cantly increased the median survival of mice (Fig. 7). Interestingly, the reduced growth of contralaterally injected B16F10 cells sug- gested the presence of a long lasting immunological memory also against non-targeted antigens. Concerning the functional state of tumor infiltrating lymphocytes (TILs), we found that all the immune therapies would enhance the percentage of activated (PD-1pos TIM- 3neg) T lymphocytes and reduce the amount of exhausted (PD-1pos TIM-3pos) cells compared to placebo. As expected, we found that PeptiCRAd monotherapy could increase the number of antigen spe- cific CD8+ T cells compared to other treatments. However, only the combination with PD-L1 blockade could significantly increase the ra- tio between activated and exhausted pentamer positive cells (p= 0.0058), suggesting that by disrupting the PD-1/PD-L1 axis we could decrease the amount of dysfunctional antigen specific T cells. We ob- served that the anatomical location deeply influenced the state of CD4+ and CD8+ T lymphocytes. In fact, TIM-3 expression was in- creased by 2 fold on TILs compared to splenic and lymphoid T cells. In the CD8+ compartment, the expression of PD-1 on the surface seemed to be restricted to the tumor micro-environment, while CD4 + T cells had a high expression of PD-1 also in lymphoid organs. Interestingly, we found that the levels of PD-1 were significantly higher on CD8+ T cells than on CD4+ T cells into the tumor micro- environment (p < 0.0001). Conclusions In conclusion, we demonstrated that the efficacy of immune check- point inhibitors might be strongly enhanced by their combination with cancer vaccines. PeptiCRAd was able to increase the number of antigen-specific T cells and PD-L1 blockade prevented their exhaus- tion, resulting in long-lasting immunological memory and increased median survival

    Development of a Robust and Scalable Synthetic Route for a Potent and Selective Isoindolinone PI3Kγ Inhibitor

    No full text
    We recently described the structure-guided optimization of a series of pyrazolopyrimidine isoindolinone PI3Kγ inhibitors, which resulted in the identification of an advanced lead compound (1) with favorable potency, selectivity, and drug-like properties. To support preclinical characterization of 1, a robust and scalable synthesis was required. Herein, we report the development of an optimized synthesis of 1, which features a scalable difluoromethylation protocol and a one-pot borylation/Suzuki–Miyaura cross-coupling reaction to access the biaryl core of the molecule. A method was developed for the efficient removal of residual palladium following Pd-catalyzed cross-coupling, which provided access to 1 in high purity without the use of any chromatographic purifications. A comprehensive investigation of solid-state polymorphism identified a thermodynamically stable crystalline form of 1, greater than 200 g of which were prepared using our optimized synthesis

    Characterization of Pharmacologic and Pharmacokinetic Properties of CCX168, a Potent and Selective Orally Administered Complement 5a Receptor Inhibitor, Based on Preclinical Evaluation and Randomized Phase 1 Clinical Study.

    No full text
    The complement 5a receptor has been an attractive therapeutic target for many autoimmune and inflammatory disorders. However, development of a selective and potent C5aR antagonist has been challenging. Here we describe the characterization of CCX168 (avacopan), an orally administered selective and potent C5aR inhibitor. CCX168 blocked the C5a binding, C5a-mediated migration, calcium mobilization, and CD11b upregulation in U937 cells as well as in freshly isolated human neutrophils. CCX168 retains high potency when present in human blood. A transgenic human C5aR knock-in mouse model allowed comparison of the in vitro and in vivo efficacy of the molecule. CCX168 effectively blocked migration in in vitro and ex vivo chemotaxis assays, and it blocked the C5a-mediated neutrophil vascular endothelial margination. CCX168 was effective in migration and neutrophil margination assays in cynomolgus monkeys. This thorough in vitro and preclinical characterization enabled progression of CCX168 into the clinic and testing of its safety, tolerability, pharmacokinetic, and pharmacodynamic profiles in a Phase 1 clinical trial in 48 healthy volunteers. CCX168 was shown to be well tolerated across a broad dose range (1 to 100 mg) and it showed dose-dependent pharmacokinetics. An oral dose of 30 mg CCX168 given twice daily blocked the C5a-induced upregulation of CD11b in circulating neutrophils by 94% or greater throughout the entire day, demonstrating essentially complete target coverage. This dose regimen is being tested in clinical trials in patients with anti-neutrophil cytoplasmic antibody-associated vasculitis. Trial Registration ISRCTN registry with trial ID ISRCTN13564773

    <i>In vitro</i> Characterization of CCX168 Using Freshly-Isolated Human Neutrophils or Human Whole Blood.

    No full text
    <p>(A) Sequential intracellular calcium release in human neutrophils in response to C5a (100 nM), CXCL8 (100 nM) or ionomycin (1 μg/mL), in the presence (blue line) or absence (black line) of CCX168 (10 μM). CCX168 blocked calcium release induced by C5a but not by the CXCR1 ligand CXCL8. (B) Binding of [<sup>125</sup>I]-C5a to human neutrophils in the presence of a range of concentrations of CCX168. CCX168 inhibited [<sup>125</sup>I]-C5a binding with a potency (IC<sub>50</sub> value) of 0.2 nM. Each data point represents the mean of 4 replicates ± standard error, and the experiment was repeated 2 separate times. (C) C5a-induced chemotaxis of leukocytes in human whole blood, in the presence of vehicle control (■), and 25 nM (○) or 250 nM (●) CCX168. CCX168 inhibited leukocyte chemotaxis in a dose-dependent manner. Each data point represents the mean of 8 replicates ± standard error, and the experiment was repeated 5 separate times. (D) C5a-induced upregulation of CD11b on the surface of neutrophils in human whole blood in the presence of vehicle control (○) or 50 nM CCX168 (●). CCX168 inhibited CD11b upregulation. Each data point represents the mean of 4 replicates ± standard error, and the experiment was repeated 4 separate times. (E) C5a-induced oxidative burst in isolated human neutrophils in the presence of vehicle control (red histogram) or CCX168 (100 nM, blue histogram). The empty histograms represent untreated neutrophils (i.e., no C5a) in the presence of vehicle control (solid black line) or CCX168 (dotted black line). CCX168 blocked the C5a-induced oxidative burst but did not affect untreated neutrophils. The experiment was repeated 3 times. (F) Chemotaxis of leukocytes in human whole blood towards synovial fluid in the presence of vehicle control or CCX168 (100 nM). Experiments using synovial fluid from a patient with rheumatoid arthritis (RA) or osteoarthritis (OA) are shown. CCX168 inhibited leukocyte chemotaxis induced by each of these samples. Each bar represents the mean of 8 replicates ± standard error, and the experiment was repeated two times. **p<0.01 based on Student’s t-test.</p

    Biological Effects of CCX168 in Cynomolgus Monkeys.

    No full text
    <p>(A) <i>In vitro</i> C5a-induced chemotaxis of freshly-isolated neutrophils from cynomolgus monkeys, conducted in 100% cynomolgus plasma containing vehicle (□) or 50 nM CCX168 (●). CCX168 inhibited neutrophil chemotaxis (A<sub>2</sub> = 3.0 nM). Each data point represents the mean of 8 replicates ± standard error, and the study was repeated 2 separate times. (B) <i>In vivo</i> effect of CCX168 in the C5a-induced neutropenia model in monkeys. The percent change in the number of neutrophils in the blood collected after C5a injection, relative to the sample collected prior to C5a injection, is shown (4 monkeys per group). Above each bar is the average concentration of CCX168 in the pre-injection blood samples. CCX168 inhibited, in a dose-dependent manner, the depletion of blood neutrophils caused by intravenous administration of C5a. **p<0.01 based on Student’s t-test.</p

    Biological Effects of CCX168 on Transgenic Human C5aR Knock-in Mice.

    No full text
    <p>(A) C5a-induced upregulation of CD11b on the surface of neutrophils in whole blood from human C5aR knock-in mice, after addition of vehicle (■), 10 nM (○), or 100 nM (●) CCX168. CCX168 inhibited CD11b upregulation in a dose-dependent manner. Each data point represents the mean of 3 replicates ± standard error; the study was repeated 3 separate times. (B) C5a-induced upregulation of CD11b on the surface of neutrophils in whole blood from human C5aR knock-in mice 1 hour after oral dosing with vehicle (●), 0.075 mg/kg (□), or 0.15 mg/kg (■) CCX168. Neutrophil CD11b upregulation was diminished in blood from CCX168-treated mice. Plasma concentrations of CCX168 are indicated. (C) Schematic of the C5a-induced leukopenia study in human C5aR knock-in mice. One hour following oral administration of CCX168, C5a (20 μg/kg) was administered intravenously, with blood drawn immediately before and 1 minute after C5a injection. Blood samples were analyzed for leukocyte numbers. (D) Effect of CCX168 in the C5a-induced leukopenia study in hC5aR knock-in mice. The percent change in the number of leukocytes in the blood sample collected after C5a injection, relative to the sample collected prior to C5a injection, is shown for each group (4 mice per group). Above each bar is the average concentration of CCX168 in the pre-injection blood samples. CCX168 inhibited the depletion of blood leukocytes caused by intravenous administration of C5a. *p<0.05, **p<0.01 based on Student’s t-test.</p

    The Complement Cascade Showing the Point of Intervention of CCX168.

    No full text
    <p>The complement cascade can be activated by the classical, lectin, or alternative pathways and leads to the formation of C3a, C3b, C5b-9 (terminal complement complex) and C5a. During the amplification loop, full length C3 is cleaved to form C3a and C3b; C3aR is the G protein coupled expressed receptor that responds to C3a, while C3b covalently binds to foreign surfaces and aids in phagocytosis and clearance. C5 convertase is formed during the amplification loop, leading to the cleavage of full length C5 at a specific arginine-leucine bond to form C5a and C5b. C5b associates with complement components C6, C7, C8, and C9 to form the terminal complement complex or membrane attack complex (MAC), typically on the surface of pathogenic bacterial cells. C5aR (CD88) is the G protein coupled receptor expressed by innate immune cells, such as neutrophils, that responds to C5a (12 kDa), a potent pro-inflammatory mediator [<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0164646#pone.0164646.ref002" target="_blank">2</a>,<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0164646#pone.0164646.ref011" target="_blank">11</a>]. C5a rapidly induces expression of adhesion molecules on the cell surface of innate immune cells, such as neutrophils, and induces the directed migration, or chemotaxis, of these cells. C5a also mediates inflammation by stimulating vascular permeability, neutrophil degranulation, and release of lysosomal proteases and oxidative free radicals [<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0164646#pone.0164646.ref008" target="_blank">8</a>]. C5a is a transient molecule, being rapidly degraded of its carboxy terminal arginine [<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0164646#pone.0164646.ref012" target="_blank">12</a>], and hence losing about 10-fold activity on C5aR, and then being internalized and degraded via C5L2, a 7 trans-membrane receptor that has an anti-inflammatory role [<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0164646#pone.0164646.ref013" target="_blank">13</a>–<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0164646#pone.0164646.ref017" target="_blank">17</a>]. CCX168 is a small molecule antagonist of C5aR that selectively and competitively binds to this receptor.</p

    <i>In vitro</i> Characterization of CCX168 Using C5aR-Expressing U937 Cells.

    No full text
    <p>(A) CCX168 inhibition of [<sup>125</sup>I]-C5a binding to U937 cells with a potency (IC<sub>50</sub> value) of 0.1 nM; each data point is the mean of 4 replicates ± standard error; study repeated 5 separate times, representative experiment shown. (B) Effects of vehicle control (■) and 1 nM (○) or 10 nM (●) CCX168 on C5a-mediated chemotaxis of U937 cells in buffer; each data point is the mean of 8 replicates ± standard error; study repeated 5 separate times, representative experiment shown. (C) C5a-mediated chemotaxis of U937 cells in the absence (●) or presence (■) of 1 μM CCX168, as well as following 1 μM CCX168 / 3x wash (□) and 1 μM CCX168 / 6x wash (Δ) treatments; each data point is the mean of 8 replicates ± standard error; study repeated 2 separate times, representative experiment shown. (D) C5a-induced intracellular calcium release in U937 cells, as measured by FLIPR, in the presence of vehicle control (●) and various concentrations of CCX168, 1 nM (○), 10 nM (■), or 100 nM (▲); each data point is the mean of 4 replicates ± standard error; study repeated 2 separate times, representative experiment shown. (E) C5a-mediated chemotaxis of U937 cells in 100% human plasma in the presence of vehicle control (■) and 1 nM (○) or 10 nM (●) CCX168; each data point is the mean of 8 replicates ± standard error; study repeated 2 separate times, representative experiment shown. (F) Inhibition by CCX168 of U937 cell chemotaxis towards 0.1 nM C5a in the presence (□) or absence (●) of α1-acid glycoprotein (AGP, 5 mg/mL) in buffer containing 5% human serum albumin (HSA); each data point is the mean of 8 replicates ± standard error; study repeated 2 separate times, representative experiment shown.</p
    corecore