Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 38
1.
Clin Transl Sci ; 15(1): 55-62, 2022 01.
Article En | MEDLINE | ID: mdl-33742767

Inhibitor of apoptosis proteins (IAPs) regulate apoptosis and modulate NF-κB signaling thereby driving expression of genes involved in immune/inflammatory responses. The orally available IAP antagonist Debio 1143 has potential to enhance tumor response to chemoradiotherapy and/or immunotherapy. Patients with pre-operative squamous cell carcinomas of the head and neck (SCCHN) received: Debio 1143 monotherapy (200 mg/day [D]1-15 +/- 2); Debio 1143 (200 mg/day D1-15 +/- 2) plus cisplatin (40 mg/m2 D 1 and 8); cisplatin alone (40 mg/m2 D 1 and 8; EudraCT: 2014-004655-31). Pharmacokinetic/pharmacodynamic effects were assessed in plasma and resected tumors. Primary end point; effect of Debio 1143 on cellular IAP-1 (cIAP-1). Levels of cIAP-1/-2, X-linked inhibitor of apoptosis protein (XIAP), tumor infiltrating lymphocytes (TILs), including CD8+ T cells, programmed cell death protein 1 (PD-1), PD-ligand 1 (PD-L1), and gene expression were also analyzed. Twenty-three of 26 patients completed treatment. In the Debio 1143 monotherapy cohort (n = 13), mean tumor concentrations of Debio 1143 were 18-fold (maximum 55.2-fold) greater than in plasma, exceeding the half-maximal inhibitory concentration for cIAPs and XIAP by 100 to 1000-fold, with significant engagement/degradation of cIAP-1 (p < 0.05). Overall, levels of CD8+ TILs, PD-1, and PD-L1 positive immune cells increased significantly (p < 0.05) following Debio 1143 treatment. Changes were observed in the expression of genes related to NF-κB signaling. Treatments were well-tolerated. Debio 1143 penetrated SCCHN tumors, engaged cIAP-1, and induced immune inflammatory changes in the tumor microenvironment. Based on the mode of action demonstrated here and in previous studies, these data support future combinations of Debio 1143 with immune-checkpoint agents.


Inhibitor of Apoptosis Proteins/pharmacology , Inhibitor of Apoptosis Proteins/pharmacokinetics , Squamous Cell Carcinoma of Head and Neck/drug therapy , Tumor Microenvironment/drug effects , Clinical Trials as Topic , Cohort Studies , Humans , Inhibitor of Apoptosis Proteins/administration & dosage , Pharmacogenetics
2.
Clin Cancer Res ; 26(24): 6429-6436, 2020 12 15.
Article En | MEDLINE | ID: mdl-32994295

PURPOSE: Debio 1143 is an oral antagonist of inhibitor of apoptosis proteins, which enhances tumor response with concomitant chemoradiotherapy. Addition of Debio 1143 to cisplatin-based chemoradiotherapy in locally advanced squamous cell carcinomas of the head and neck (LA-SCCHN) was evaluated in a phase I/II study to determine the MTD and recommended phase II dose (RP2D). Here, phase I results are reported. PATIENTS AND METHODS: Treatment-naïve patients with LA-SCCHN (stages III/IVA/IVB) received Debio 1143 (100, 200, 300 mg/day), for 14 days every 3 weeks, with cisplatin (100 mg/m², every 3 weeks), for three cycles, and concomitant conventional fractionation radiotherapy (70 Gy/7 weeks). Dose-limiting toxicity (DLT) was evaluated over 9 weeks using continual reassessment. RESULTS: Fourteen patients were treated/evaluable for DLT. Median age was 64.5 years, and all patients were current/former smokers. Primary tumors were hypopharynx, oropharynx (all human papillomavirus/p16 negative), larynx, and oral cavity. Two of six patients at 200 mg/day had DLT (grade 3 tubular necrosis, grade 3 aspartate aminotransferase/alanine aminotransferase increase, grade 4 febrile neutropenia, and grade 3 lipase increase), which was considered the MTD and RP2D. Common grade 3-4 adverse events were dysphagia (36%) and mucositis (29%). Laboratory abnormalities were frequent and generally mild, including anemia, white blood cell decrease, and increased creatinine. Addition of Debio 1143 did not compromise chemotherapy administration. Overall locoregional control rate at 18 months was 85%. Overall response rate was 85%, including 69% complete responses. Progression-free survival rate at 24 months was 74%. CONCLUSIONS: The RP2D of Debio 1143 is 200 mg/day for 14 days, every 3 weeks, when combined with concomitant high-dose cisplatin chemoradiotherapy in LA-SCCHN. Debio 1143 addition to chemoradiotherapy was safe and manageable. Preliminary efficacy is encouraging and supports further development.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemoradiotherapy/methods , Head and Neck Neoplasms/therapy , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Squamous Cell Carcinoma of Head and Neck/therapy , Adolescent , Adult , Aged , Azocines/administration & dosage , Benzhydryl Compounds/administration & dosage , Cisplatin/administration & dosage , Female , Follow-Up Studies , Head and Neck Neoplasms/pathology , Humans , Male , Maximum Tolerated Dose , Middle Aged , Prognosis , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck/pathology , Young Adult
3.
PLoS One ; 15(1): e0227715, 2020.
Article En | MEDLINE | ID: mdl-31978106

The immune checkpoint programmed cell death protein 1 (PD-1) plays a major role in T cell exhaustion in cancer and chronic HIV infection. The inhibitor of apoptosis protein antagonist Debio 1143 (D1143) enhances tumor cell death and synergizes with anti-PD-1 agents to promote tumor immunity and displayed HIV latency reversal activity in vitro. We asked in this study whether D1143 would stimulate the potency of an anti-human PD-1 monoclonal antibody (mAb) to reduce HIV loads in humanized mice. Anti-PD-1 mAb treatment decreased PD-1+ CD8+ cell population by 32.3% after interruption of four weeks treatment, and D1143 co-treatment further reduced it from 32.3 to 73%. Anti-PD-1 mAb administration reduced HIV load in blood by 94%, and addition of D1143 further enhanced this reduction from 94 to 97%. D1143 also more profoundly promoted with the anti-PD-1-mediated reduction of HIV loads in all tissues analyzed including spleen (71 to 96.4%), lymph nodes (64.3 to 80%), liver (64.2 to 94.4), lung (64.3 to 80.1%) and thymic organoid (78.2 to 98.2%), achieving a >5 log reduction of HIV loads in CD4+ cells isolated from tissues 2 weeks after drug treatment interruption. Ex vivo anti-CD3/CD28 stimulation increased the ability to activate exhausted CD8+ T cells in infected mice having received in vivo anti-PD-1 treatment by 7.9-fold (5 to 39.6%), and an additional increase by 1.7-fold upon D1143 co-treatment (39.6 to 67.3%). These findings demonstrate for the first time that an inhibitor of apoptosis protein antagonist enhances in a statistically manner the effects of an immune check point inhibitor on antiviral immunity and on HIV load reduction in tissues of humanized mice, suggesting that the combination of two distinct classes of immunomodulatory agents constitutes a promising anti-HIV immunotherapeutic approach.


Antibodies, Monoclonal/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , HIV Infections/drug therapy , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Azocines/therapeutic use , Benzhydryl Compounds/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Synergism , Drug Therapy, Combination , Female , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , HIV-1/isolation & purification , Humans , Mice , Mice, Transgenic , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Viral Load/drug effects , Viral Load/immunology
4.
PLoS One ; 14(2): e0211746, 2019.
Article En | MEDLINE | ID: mdl-30716099

Antiretroviral therapy (ART) suppresses HIV replication, but does not cure the infection because replication-competent virus persists within latently infected CD4+ T cells throughout years of therapy. These reservoirs contain integrated HIV-1 genomes and can resupply active virus. Thus, the development of strategies to eliminate the reservoir of latently infected cells is a research priority of global significance. In this study, we tested efficacy of a new inhibitor of apoptosis protein antagonist (IAPa) called Debio 1143 at reversing HIV latency and investigated its mechanisms of action. Debio 1143 activates HIV transcription via NF-kB signaling by degrading the ubiquitin ligase baculoviral IAP repeat-containing 2 (BIRC2), a repressor of the non-canonical NF-kB pathway. Debio 1143-induced BIRC2 degradation results in the accumulation of NF-κB-inducing kinase (NIK) and proteolytic cleavage of p100 into p52, leading to nuclear translocation of p52 and RELB. Debio 1143 greatly enhances the binding of RELB to the HIV-1 LTR. These data indicate that Debio 1143 activates the non-canonical NF-kB signaling pathway by promoting the binding of RELB:p52 complexes to the HIV-1 LTR, resulting in the activation of the LTR-dependent HIV-1 transcription. Importantly, Debio 1143 reverses viral latency in HIV-1 latent T cell lines. Using knockdown (siRNA BIRC2), knockout (CRIPSR NIK) and proteasome machinery neutralization (MG132) approaches, we found that Debio 1143-mediated HIV latency reversal is BIRC2 degradation- and NIK stabilization-dependent. Debio 1143 also reverses HIV-1 latency in resting CD4+ T cells derived from ART-treated patients or HIV-1-infected humanized mice under ART. Interestingly, daily oral administration of Debio 1143 in cancer patients at well-tolerated doses elicited BIRC2 target engagement in PBMCs and induced a moderate increase in cytokines and chemokines mechanistically related to NF-kB signaling. In conclusion, we provide strong evidences that the IAPa Debio 1143, by initially activating the non-canonical NF-kB signaling and subsequently reactivating HIV-1 transcription, represents a new attractive viral latency reversal agent (LRA).


Anti-HIV Agents/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , CD4-Positive T-Lymphocytes/metabolism , HIV-1/physiology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Virus Latency/drug effects , Animals , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Mice , NF-kappa B p52 Subunit/genetics , NF-kappa B p52 Subunit/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factor RelB/genetics , Transcription Factor RelB/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
5.
Clin Cancer Res ; 25(3): 1113-1124, 2019 02 01.
Article En | MEDLINE | ID: mdl-30352911

PURPOSE: Adaptive antitumor immunity following ablative radiotherapy (ART) is attenuated by host myeloid-derived suppressor cell (MDSC), tumor-associated macrophage (TAM), and regulatory T-cell (Treg) infiltrates. We hypothesized treatment with ART and a secondary mitochondrial-derived activators of caspase (SMAC) mimetic could reverse the immunosuppressive lung cancer microenvironment to favor adaptive immunity. EXPERIMENTAL DESIGN: To evaluate for synergy between ART and the SMAC mimetic Debio 1143 and the dependence upon CD8+ T cells and TNFα, we used LLC-OVA syngeneic mouse model of lung cancer and treated them with Debio 1143 and/or ART (30 Gy) with or without anti-CD8, anti-TNFα, or anti-IFNγ antibodies. Tumor-infiltrating OVA-specific CD8+ T cells, Tc1 effector cells, MDSCs, TAMs, and Tregs, were quantified by flow cytometry. Tc1-promoting cytokines TNFα, IFNγ, and IL1ß and the immunosuppressive IL10 and Arg-1 within LLC-OVA tumor tissue or mouse serum were measured by RT-PCR and ELISA. RESULTS: ART delayed tumor growth, and the addition of Debio 1143 greatly enhanced its efficacy, which included several complete responses. These complete responders rejected an LLC-OVA tumor rechallenge. ART and Debio 1143 synergistically induced a tumor-specific, Tc1 cellular and cytokine response while eliminating immunosuppressive cells and cytokines from the tumor microenvironment. Depletion of CD8+ cells, TNFα, and IFNγ with blocking antibody abrogated synergy between ART and Debio 1143 and partially restored tumor-infiltrating MDSCs. CONCLUSIONS: Debio 1143 augments the tumor-specific adaptive immunity induced by ART, while reversing host immunosuppressive cell infiltrates in the tumor microenvironment in a TNFα, IFNγ, and CD8+ T-cell-dependent manner. This provides a novel strategy to enhance the immunogenicity of ART.


Apoptosis Regulatory Proteins/metabolism , Azocines/therapeutic use , Benzhydryl Compounds/therapeutic use , Biomimetic Materials/therapeutic use , Carcinoma, Lewis Lung/therapy , Lung Neoplasms/therapy , Mitochondrial Proteins/metabolism , Radiotherapy/methods , Animals , Apoptosis/drug effects , Apoptosis/immunology , Apoptosis/radiation effects , Azocines/immunology , Benzhydryl Compounds/immunology , Biomimetic Materials/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Combined Modality Therapy , Cytokines/blood , Cytokines/genetics , Cytokines/immunology , Female , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/radiation effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
6.
Sci Rep ; 8(1): 17862, 2018 12 14.
Article En | MEDLINE | ID: mdl-30552344

The poor prognosis of ovarian cancer (it is the leading cause of death from gynecological cancers) is mainly due to the acquisition of resistance to carboplatin. Among the possible resistance pathways, resistance to apoptosis and especially the overexpression of inhibitor of apoptosis proteins (IAP) cIAP1 and X-linked IAP (XIAP), have been implicated. DEBIO 1143, a SMAC (second mitochondria-derived activator of caspase) mimetic, belongs to a new class of targeted agents currently being evaluated in clinical trials, which activate apoptotic cell death and block pro-survival signaling in cancer cells. Here, we demonstrate that DEBIO 1143 in vitro inhibits the cell viability of two carboplatin-sensitive cell lines (IGROV-1 and A2780S) as well as three carboplatin-resistant cell lines (A2780R, SKOV-3 and EFO-21). Of note, DEBIO 1143 is able to reverse resistance to carboplatin by inducing cell death either by apoptosis or necroptosis depending on the cell lines. To identify a biomarker able to predict the sensitivity of the cell lines to DEBIO 1143 treatment we analyzed the expression of the DEBIO 1143 targets cIAP1 and XIAP, and one of their downstream targets, caspase 9. These proteins did not constitute a marker of DEBIO 1143 sensitivity/resistance. Importantly, we confirmed these findings in vivo in SKOV-3 xenograft models where DEBIO 1143 highly potentiated carboplatin treatment.


Antineoplastic Agents/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , Carboplatin/pharmacology , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Azocines/administration & dosage , Benzhydryl Compounds/administration & dosage , Carboplatin/administration & dosage , Caspase 9/analysis , Cell Line, Tumor , Disease Models, Animal , Female , Heterografts , Humans , Inhibitor of Apoptosis Proteins/analysis , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/drug therapy , Treatment Outcome , X-Linked Inhibitor of Apoptosis Protein/analysis
7.
Contrast Media Mol Imaging ; 2018: 8494031, 2018.
Article En | MEDLINE | ID: mdl-30627061

Background: Debio 1143, a potent orally available SMAC mimetic, targets inhibitors of apoptosis proteins (IAPs) members and is currently in clinical trials. In this study, nuclear imaging evaluated the effects of Debio 1143 on tumor cell death and metabolism in a triple-negative breast cancer (TNBC) cell line (MDA-MB-231)-based animal model. Methods: Apoptosis induced by Debio 1143 was assessed by FACS (caspase-3, annexin 5 (A5)), binding of 99mTc-HYNIC-Annexin V, and a cell proliferation assay. 99mTc-HYNIC-Annexin V SPECT and [18F]-FDG PET were also performed in mice xenografted with MDA-MB-231 cells. Results: Debio 1143 induced early apoptosis both in vitro and in vivo 6 h after treatment. Debio 1143 inhibited tumor growth, which was associated with a decreased tumor [18F]-FDG uptake when measured during treatment. Conclusions: This imaging study combining SPECT and PET showed the early proapoptotic effects of Debio 1143 resulting in a robust antitumor activity in a preclinical TNBC model. These imaging biomarkers represent valuable noninvasive tools for translational and clinical research in TNBC.


Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Multimodal Imaging/methods , Radiopharmaceuticals/chemistry , Triple Negative Breast Neoplasms/diagnostic imaging , Animals , Apoptosis/drug effects , Biomarkers , Female , Heterografts , Humans , Magnetic Resonance Imaging/methods , Mice , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals/pharmacology , Tomography, Emission-Computed, Single-Photon , Translational Research, Biomedical , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
8.
Infect Genet Evol ; 44: 169-181, 2016 10.
Article En | MEDLINE | ID: mdl-27374748

Major advances have revolutionized the HCV antiviral treatment field, with interferon-free combinations of direct-acting antivirals (DAAs) resulting into success rates of >90% for all HCV genotypes. Nevertheless, viral eradication at a global level stills remains challenging, stimulating the continued search for new affordable pan-genotypic drugs. To overcome selection of drug resistant variants, targeting host proteins can be an attractive mechanism of action. Alisporivir (Debio 025) is a potent pan-genotypic host-targeting antiviral agent, acting on cyclophilin A, which is necessary for HCV replication. The efficacy and safety of three different oral doses of alisporivir in combination with pegylated interferon-α2a given over a period of four weeks, was investigated in a randomized, double-blind and placebo-controlled phase IIa clinical trial, in 90 treatment-naïve subjects infected with chronic hepatitis C, wherefrom 58 HCV1b samples were selected for genetic sequencing purposes. Sequencing results were used to study the HCV genome for amino acid changes potentially related with selective pressure and resistance to alisporivir. By comparing baseline and on-treatment sequences, a large variation in proportion of amino acid changes was detected in all treatment arms. The NS5A variant D320E, which was previously identified during in vitro resistance selection and resulted in 3.6-fold reduced alisporivir susceptibility, emerged in two subjects in the alisporivir monotherapy arm. However, emergence of D320E appeared to be associated only with concurrent viral load rebound in one subject with 0.8log10IU/ml increase in HCV RNA. In general, for all datasets, low numbers of positions under positive selective pressure were observed, with no significant differences between naïve and treated sequences. Additionally, incomplete sequence information for some of the 22 patients and the low number of individuals per treatment arm, is limiting the power to assess the association of alisporivir or interferon treatment with the observed amino acid changes.


Cyclosporine/therapeutic use , Hepacivirus/drug effects , Hepacivirus/genetics , Hepatitis C, Chronic/drug therapy , Antiviral Agents/therapeutic use , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Genome, Viral , Hepatitis C, Chronic/virology , Humans , Interferon-alpha/therapeutic use , Phylogeny , Polyethylene Glycols/therapeutic use , Recombinant Proteins/therapeutic use , Viral Load/drug effects , Viral Nonstructural Proteins/genetics
9.
Antimicrob Agents Chemother ; 60(7): 4264-73, 2016 07.
Article En | MEDLINE | ID: mdl-27161626

Broad-spectrum antibiotic therapy decimates the gut microbiome, resulting in a variety of negative health consequences. Debio 1452 is a staphylococcus-selective enoyl-acyl carrier protein reductase (FabI) inhibitor under clinical development and was used to determine whether treatment with pathogen-selective antibiotics would minimize disturbance to the microbiome. The effect of oral Debio 1452 on the microbiota of mice was compared to the effects of four commonly used broad-spectrum oral antibiotics. During the 10 days of oral Debio 1452 treatment, there was minimal disturbance to the gut bacterial abundance and composition, with only the unclassified S24-7 taxon reduced at days 6 and 10. In comparison, broad-spectrum oral antibiotics caused ∼100- to 4,000-fold decreases in gut bacterial abundance and severely altered the microbial composition. The gut bacterial abundance and composition of Debio 1452-treated mice were indistinguishable from those of untreated mice 2 days after the antibiotic treatment was stopped. In contrast, the bacterial abundance in broad-spectrum-antibiotic-treated mice took up to 7 days to recover, and the gut composition of the broad-spectrum-antibiotic-treated mice remained different from that of the control group 20 days after the cessation of antibiotic treatment. These results illustrate that a pathogen-selective approach to antibiotic development will minimize disturbance to the gut microbiome.


Anti-Bacterial Agents/pharmacology , Animals , Feces/microbiology , Female , Gastrointestinal Microbiome/drug effects , Mice , Mice, Inbred C57BL , Microbiota/drug effects , Microbiota/genetics , RNA, Ribosomal, 16S/genetics
10.
Oncotarget ; 6(35): 37410-25, 2015 Nov 10.
Article En | MEDLINE | ID: mdl-26485762

Targeting anti-apoptotic proteins can sensitize tumor cells to conventional chemotherapies or other targeted agents. Antagonizing the Inhibitor of Apoptosis Proteins (IAPs) with mimetics of the pro-apoptotic protein SMAC is one such approach. We used sensitization compound screening to uncover possible agents with the potential to further sensitize lung adenocarcinoma cells to the SMAC mimetic Debio 1143. Several compounds in combination with Debio 1143, including taxanes, topoisomerase inhibitors, and bromodomain inhibitors, super-additively inhibited growth and clonogenicity of lung adenocarcinoma cells. Co-treatment with Debio 1143 and the bromodomain inhibitor JQ1 suppresses the expression of c-IAP1, c-IAP2, and XIAP. Non-canonical NF-κB signaling is also activated following Debio 1143 treatment, and Debio 1143 induces the formation of the ripoptosome in Debio 1143-sensitive cell lines. Sensitivity to Debio 1143 and JQ1 co-treatment was associated with baseline caspase-8 expression. In vivo treatment of lung adenocarcinoma xenografts with Debio 1143 in combination with JQ1 or docetaxel reduced tumor volume more than either single agent alone. As Debio 1143-containing combinations effectively inhibited both in vitro and in vivo growth of lung adenocarcinoma cells, these data provide a rationale for Debio 1143 combinations currently being evaluated in ongoing clinical trials and suggest potential utility of other combinations identified here.


Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azepines/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , Camptothecin/analogs & derivatives , Cell Proliferation/drug effects , Lung Neoplasms/drug therapy , Paclitaxel/pharmacology , Taxoids/pharmacology , Topoisomerase Inhibitors/pharmacology , Triazoles/pharmacology , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Camptothecin/pharmacology , Cell Line, Tumor , Docetaxel , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Irinotecan , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Radiother Oncol ; 116(3): 495-503, 2015 Sep.
Article En | MEDLINE | ID: mdl-26096848

BACKGROUND AND PURPOSE: Second mitochondria-derived activator of caspase (SMAC)-mimetics are a new class of targeted drugs that specifically induce apoptotic cancer cell death and block pro-survival signaling by antagonizing selected members of the inhibitor of apoptosis protein (IAP) family. MATERIAL AND METHODS: The present study was designed to investigate the radiosensitizing effect and optimal sequence of administration of the novel SMAC-mimetic Debio 1143 in vitro and in vivo. Apoptosis, alteration of DNA damage repair (DDR), and tumor necrosis factor-alpha (TNF-α) signaling were examined. RESULTS: In vitro, Debio 1143 displayed anti-proliferative activity and enhanced intrinsic radiation sensitivity in 5/6 head and neck squamous cell carcinoma (HNSCC) cell lines in a synergistic manner. In vivo, Debio 1143 dose-dependently radio-sensitized FaDu and SQ20B xenografts, resulting in complete tumor regression in 8/10 FaDu-xenografted mice at the high dose level. At the molecular level, Debio 1143 combined with radiotherapy (RT) induced enhancement of caspase-3 activity, increase in Annexin V-positive cells and karyopyknosis, and increase in TNF-α mRNA levels. Finally, in a neutralization experiment using a TNF-α-blocking antibody and a caspase inhibitor, it was shown that the radiosensitizing effect of Debio 1143 is mediated by caspases and TNF-α. CONCLUSIONS: These results demonstrate that the novel SMAC-mimetic Debio 1143 is a radiosensitizing agent that is worthy of further investigation in clinical trials in combination with radiotherapy.


Antineoplastic Agents/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/therapy , Radiation-Sensitizing Agents/pharmacology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Caspase 3/metabolism , Cell Death/drug effects , Cell Line, Tumor , Chemoradiotherapy/methods , Female , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/pharmacology , Mice, Inbred Strains , Mitochondrial Proteins/pharmacology , Neoplasm Transplantation , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck , Transplantation, Heterologous , Xenograft Model Antitumor Assays/methods
12.
Clin Lymphoma Myeloma Leuk ; 15(7): 443-9, 2015 Jul.
Article En | MEDLINE | ID: mdl-25842225

BACKGROUND: Treatment of acute myeloid leukemia (AML) remains difficult owing to the development of treatment resistance, which might be overcome through antagonists of inhibitors of apoptosis proteins (IAPs). PATIENTS AND METHODS: The present multicenter, open-label, dose-escalation study aimed to evaluate the tolerability, pharmacokinetics (PK), pharmacodynamics (PD), and efficacy of Debio1143 (formerly AT-406), a new IAP antagonist, when given along with a standard "7 plus 3 regimen" of daunorubicin and cytarabine to poor-risk patients with AML during the induction cycle. Consecutive patient cohorts received once-daily 100, 200, 300, or 400 mg of oral Debio1143 on treatment days 1 to 5. Blood samples were collected regularly until hematologic recovery or response was documented. Bone marrow samples were collected on days 0, 14, and 29 and PK and PD samples on days 1, 3, 5, 8, and 10 and 1, 2, and 8, respectively. RESULTS: Of the 29 enrolled patients, 23 completed the study. The most common adverse events of any grade deemed related to treatment were nausea (31% of patients), diarrhea (14%), and febrile neutropenia (14%). Exposure exceeded dose proportionality, without accumulation over 5 days. Inhibition of cellular IAP1 was detectable in the CD34/CD117(+) cells and blasts. A total of 11 patients (38%) achieved complete remission, most in the 100-mg dose cohort. Of these, 6 (56%) developed a relapse within the study period. The patients with a response more frequently showed plasma increases of tumor necrosis factor-α and interleukin-8 after the first dose of Debio1143. CONCLUSION: Debio1143 ≤ 400 mg/d showed good tolerability in combination with daunorubicin and cytarabine. Additional studies in subsets of patients with AML are warranted.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azocines/administration & dosage , Benzhydryl Compounds/administration & dosage , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Administration, Oral , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cytarabine/administration & dosage , Cytokines/blood , Daunorubicin/administration & dosage , Dose-Response Relationship, Drug , Female , Humans , Leukemia, Myeloid, Acute/blood , Male , Middle Aged
13.
Cancer Chemother Pharmacol ; 75(4): 851-9, 2015 Apr.
Article En | MEDLINE | ID: mdl-25716544

PURPOSE: To assess safety/tolerability, pharmacokinetics (PK), pharmacodynamics (PD), and antitumor activity of DEBIO1143, an antagonist of inhibitor apoptosis proteins. METHODS: This first-in-man study in patients with advanced cancer used an accelerated dose titration design. DEBIO1143 was given orally once daily on days 1-5 every 2 or 3 weeks until disease progressed or patients dropped out. The starting dose of 5 mg was escalated by 100% in single patients until related grade 2 toxicity occurred. This triggered expansion to cohorts of three and subsequently six patients and reduction in dose increments to 50%. Maximum tolerated dose (MTD) was exceeded when any two patients within the same cohort experienced dose-limiting toxicity (DLT). On days 1 and 5, PK and PD samples were taken. RESULTS: Thirty-one patients received doses from 5 to 900 mg. Only one DLT was reported at 180 mg. No MTD was found. Most common adverse drug reactions were fatigue (26%), nausea (23%), and vomiting (13%). Average t max and T 1/2 was about 1 and 6 h, respectively. Exposure increased proportionally with doses from 80 to 900 mg, without accumulation over 5 days. Plasma CCL2 increased at 3-6 h postdose and epithelial apoptosis marker M30 on day 5; cIAP-1 levels in PBMCs decreased at all doses >80 mg. Five patients (17%) had stable disease as the best treatment response. CONCLUSION: DEBIO1143 was well tolerated at doses up to 900 mg and elicited PD effects at doses greater 80 mg. Limited antitumor activity may suggest development rather as adjunct treatment.


Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Azocines/administration & dosage , Azocines/therapeutic use , Benzhydryl Compounds/administration & dosage , Benzhydryl Compounds/therapeutic use , Neoplasms/drug therapy , Administration, Oral , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Azocines/adverse effects , Azocines/pharmacokinetics , Benzhydryl Compounds/adverse effects , Benzhydryl Compounds/pharmacokinetics , Chemokine CCL2/blood , Dose-Response Relationship, Drug , Female , Humans , Inhibitor of Apoptosis Proteins/blood , Male , Maximum Tolerated Dose , Neoplasms/etiology , Neoplasms/pathology , Treatment Outcome
14.
Am J Cancer Res ; 4(6): 943-51, 2014.
Article En | MEDLINE | ID: mdl-25520882

Inhibitors of apoptosis (IAPs) limit the effectiveness of radiation in non-small cell lung cancer (NSCLC). Debio 1143 (D1143) is an antagonist of IAPs. The purpose of this study was to investigate the potential of D1143 as a radiosensitizer in NSCLC. MTS assays were performed in two NSCLC cell lines: HCC193 and H460. Extent of apoptotic cell death was characterized by Annexin V assay and Western blot for cleaved caspase-3, -8, and IAPs. TNF-α release was determined by ELISA. Radiosensitivities were compared with dose enhancement ratios (DERs). HCC193 cells D1143 IC50 was 1 µM. HCC193 cells demonstrated noticeable cleaved caspase-3, -8, and a decrease in IAP levels with 2.5 µM D1143; H460 cells, with 10 µM; both in a time-dependent manner. Additionally, HCC193 cells exhibited an increase in TNF-α. D1143 radiosensitized cells: HCC193, 2.5 µM D1143, 24 h incubation, DER of 2.19, p = 0.001; H460 cells, 10 µM D1143, 48 h incubation, DER of 1.29, p = 0.082. Treatment of H460 cells with radiation therapy, TNF-α, and D1143 further radiosensitized the cells (DER of 1.92, p = 0.026). D1143 significantly enhanced the radiosensitization of HCC193 and H460 cells in vitro. TNF-α contributed to the sensitization in the more sensitive cell line (HCC193). More research is warranted to test the mechanism of D1143, and to assess its potential in vivo in the clinical setting.

15.
J Virol Methods ; 209: 126-35, 2014 Dec.
Article En | MEDLINE | ID: mdl-25245140

A near-full genome genotypic assay for HCV1b was developed, which may prove useful to investigate antiviral drug resistance, given new combination therapies for HCV1 infection. The assay consists of three partially overlapping PCRs followed by Sanger population or Illumina next-generation sequencing. Seventy-seven therapy-naïve samples, spanning the entire diversity range of currently known HCV1b, were used for optimization of PCRs, of which ten were sequenced using Sanger and of these ten, four using Illumina. The median detection limits for the three regions, 5'UTR-NS2, E2-NS5A and NS4B-NS5B, were 570, 5670 and 56,670 IU/ml respectively. The number of Illumina reads mapped varied according to the software used, Segminator II being the best performing (81%). Consensus Illumina and Sanger sequencing results accord largely (0.013% major discordances). Differences were due almost exclusively to a larger number of ambiguities (presumably minority variants) scored by Illumina (1.50% minor discordances). The assay is easy to perform in an equipped laboratory; nevertheless, it was difficult to reach high sensitivity and reproducibility, due to the high genetic viral variability. This assay proved to be suitable for detecting drug resistance mutations and can also be used for epidemiological research, even though only a limited set of samples was used for validation.


Drug Resistance, Viral , Genotyping Techniques/methods , Hepacivirus/classification , Hepacivirus/genetics , Hepatitis C/virology , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Humans , Microbial Sensitivity Tests/methods , Polymerase Chain Reaction/methods , Reproducibility of Results , Sensitivity and Specificity , Sequence Analysis, DNA/methods
16.
Acta Derm Venereol ; 94(6): 672-6, 2014 Nov.
Article En | MEDLINE | ID: mdl-24604074

Debio 0932 is a novel oral heat shock protein 90 (Hsp90) inhibitor developed for anti-cancer therapy. Surprising-ly, during the first clinical trial, one psoriasis patient experienced complete remission of his skin manifestation. However, a possible therapeutic utility of Hsp90 in psoriasis has not previously been reported. The objective of the present study was to explore the ability of Debio 0932 to alleviate psoriasis in a preclinical model. A psoriasis xenograft transplantation model was employed where skin from 5 psoriasis patients was transplanted onto immunodeficient mice (8 xenografts per donor). Debio 0932 was administered perorally daily for 3 weeks and resulted in significant clinical alleviation of psoriasis by day 11 and reduced epidermal thickness evaluated post-treatment. Alleviation of psoriasis in the psoriasis xenograft transplantation model, which may be due to Hsp90's involvement in signalling pathways that are up-regulated in psoriasis, substantiates a potential role of Debio 0932 in psoriasis treatment.


Benzodioxoles/administration & dosage , Dermatologic Agents/administration & dosage , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Imidazoles/administration & dosage , Psoriasis/drug therapy , Skin Transplantation , Skin/drug effects , Administration, Oral , Adult , Aged , Animals , Disease Models, Animal , Female , HSP90 Heat-Shock Proteins/metabolism , Heterografts , Humans , Mice, SCID , Middle Aged , Psoriasis/metabolism , Psoriasis/pathology , Remission Induction , Signal Transduction/drug effects , Skin/metabolism , Skin/pathology , Time Factors
17.
Viruses ; 5(3): 981-97, 2013 Mar 22.
Article En | MEDLINE | ID: mdl-23524389

DEB025 (alisporivir) is a synthetic cyclosporine with inhibitory activity against human immunodeficiency virus type-1 (HIV-1) and hepatitis C virus (HCV). It binds to cyclophilin A (CypA) and blocks essential functions of CypA in the viral replication cycles of both viruses. DEB025 inhibits clinical HIV-1 isolates in vitro and decreases HIV-1 virus load in the majority of patients. HIV-1 isolates being naturally resistant to DEB025 have been detected in vitro and in nonresponder patients. By sequence analysis of their capsid protein (CA) region, two amino acid polymorphisms that correlated with DEB025 resistance were identified: H87Q and I91N, both located in the CypA-binding loop of the CA protein of HIV-1. The H87Q change was by far more abundant than I91N. Additional polymorphisms in the CypA-binding loop (positions 86, 91 and 96), as well as in the N-terminal loop of CA were detected in resistant isolates and are assumed to contribute to the degree of resistance. These amino acid changes may modulate the conformation of the CypA-binding loop of CA in such a way that binding and/or isomerase function of CypA are no longer necessary for virus replication. The resistant HIV-1 isolates thus are CypA-independent.


Anti-HIV Agents/pharmacology , Capsid Proteins/genetics , Drug Resistance, Viral , HIV Infections/virology , HIV-1/genetics , Polymorphism, Genetic , Amino Acid Sequence , Capsid/drug effects , Capsid/metabolism , Capsid Proteins/metabolism , Cyclosporine , HIV Infections/drug therapy , HIV-1/drug effects , HIV-1/metabolism , Humans , Molecular Sequence Data , Mutation, Missense/drug effects
18.
J Virol ; 87(3): 1454-64, 2013 Feb.
Article En | MEDLINE | ID: mdl-23152531

Virus replication strongly depends on cellular factors, in particular, on host proteins. Here we report that the replication of the arteriviruses equine arteritis virus (EAV) and porcine reproductive and respiratory syndrome virus (PRRSV) is strongly affected by low-micromolar concentrations of cyclosporine A (CsA), an inhibitor of members of the cyclophilin (Cyp) family. In infected cells, the expression of a green fluorescent protein (GFP) reporter gene inserted into the PRRSV genome was inhibited with a half-maximal inhibitory concentration (IC(50)) of 5.2 µM, whereas the GFP expression of an EAV-GFP reporter virus was inhibited with an IC(50) of 0.95 µM. Debio-064, a CsA analog that lacks its undesirable immunosuppressive properties, inhibited EAV replication with an IC(50) that was 3-fold lower than that of CsA, whereas PRRSV-GFP replication was inhibited with an IC(50) similar to that of CsA. The addition of 4 µM CsA after infection prevented viral RNA and protein synthesis in EAV-infected cells, and CsA treatment resulted in a 2.5- to 4-log-unit reduction of PRRSV or EAV infectious progeny. A complete block of EAV RNA synthesis was also observed in an in vitro assay using isolated viral replication structures. The small interfering RNA-mediated knockdown of Cyp family members revealed that EAV replication strongly depends on the expression of CypA but not CypB. Furthermore, upon fractionation of intracellular membranes in density gradients, CypA was found to cosediment with membranous EAV replication structures, which could be prevented by CsA treatment. This suggests that CypA is an essential component of the viral RNA-synthesizing machinery.


Antiviral Agents/pharmacology , Cyclophilins/antagonists & inhibitors , Cyclosporine/pharmacology , Equartevirus/physiology , Porcine respiratory and reproductive syndrome virus/physiology , Virus Replication/drug effects , Animals , Cell Line , Gene Knockdown Techniques , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Inhibitory Concentration 50 , Lepidoptera , Microbial Sensitivity Tests , RNA, Viral/biosynthesis , Viral Proteins/biosynthesis
19.
Hepatology ; 55(5): 1333-43, 2012 May.
Article En | MEDLINE | ID: mdl-22135208

UNLABELLED: Alisporivir (Debio-025) is an analogue of cyclosporine A and represents the prototype of a new class of non-immunosuppressive cyclophilin inhibitors. In vitro and in vivo studies have shown that alisporivir inhibits hepatitis C virus (HCV) replication, and ongoing clinical trials are exploring its therapeutic potential in patients with chronic hepatitis C. Recent data suggest that the antiviral effect is mediated by inhibition of cyclophilin A, which is an essential host factor in the HCV life cycle. However, alisporivir also inhibits mitochondrial permeability transition by binding to cyclophilin D. Because HCV is known to affect mitochondrial function, we explored the effect of alisporivir on HCV protein-mediated mitochondrial dysfunction. Through the use of inducible cell lines, which allow to investigate the effects of HCV polyprotein expression independent from viral RNA replication and which recapitulate the major alterations of mitochondrial bioenergetics observed in infectious cell systems, we show that alisporivir prevents HCV protein-mediated decrease of cell respiration, collapse of mitochondrial membrane potential, overproduction of reactive oxygen species and mitochondrial calcium overload. Strikingly, some of the HCV-mediated mitochondrial dysfunctions could even be rescued by alisporivir. CONCLUSION: These observations provide new insights into the pathogenesis of HCV-related liver disease and reveal an additional mechanism of action of alisporivir that is likely beneficial in the treatment of chronic hepatitis C.


Cyclosporine/pharmacology , Hepacivirus/drug effects , Mitochondria, Liver/drug effects , Virus Replication/drug effects , Antiviral Agents/pharmacology , Apoptosis/drug effects , Calcium/metabolism , Cell Respiration/drug effects , Cells, Cultured/drug effects , Cyclophilins/antagonists & inhibitors , Hepacivirus/physiology , Humans , Immunohistochemistry , Membrane Potentials , Mitochondria, Liver/metabolism , Reactive Oxygen Species/metabolism , Sensitivity and Specificity
20.
PLoS One ; 5(10): e13687, 2010 Oct 27.
Article En | MEDLINE | ID: mdl-21060866

DEB025/Debio 025 (Alisporivir) is a cyclophilin (Cyp)-binding molecule with potent anti-hepatitis C virus (HCV) activity both in vitro and in vivo. It is currently being evaluated in phase II clinical trials. DEB025 binds to CypA, a peptidyl-prolyl cis-trans isomerase which is a crucial cofactor for HCV replication. Here we report that it was very difficult to select resistant replicons (genotype 1b) to DEB025, requiring an average of 20 weeks (four independent experiments), compared to the typically <2 weeks with protease or polymerase inhibitors. This indicates a high genetic barrier to resistance for DEB025. Mutation D320E in NS5A was the only mutation consistently selected in the replicon genome. This mutation alone conferred a low-level (3.9-fold) resistance. Replacing the NS5A gene (but not the NS5B gene) from the wild type (WT) genome with the corresponding sequence from the DEB025(res) replicon resulted in transfer of resistance. Cross-resistance with cyclosporine A (CsA) was observed, whereas NS3 protease and NS5B polymerase inhibitors retained WT-activity against DEB025(res) replicons. Unlike WT, DEB025(res) replicon replicated efficiently in CypA knock down cells. However, DEB025 disrupted the interaction between CypA and NS5A regardless of whether the NS5A protein was derived from WT or DEB025(res) replicon. NMR titration experiments with peptides derived from the WT or the DEB025(res) domain II of NS5A corroborated this observation in a quantitative manner. Interestingly, comparative NMR studies on two 20-mer NS5A peptides that contain D320 or E320 revealed a shift in population between the major and minor conformers. These data suggest that D320E conferred low-level resistance to DEB025 probably by reducing the need for CypA-dependent isomerisation of NS5A. Prolonged DEB025 treatment and multiple genotypic changes may be necessary to generate significant resistance to DEB025, underlying the high barrier to resistance.


Antiviral Agents/pharmacology , Cyclophilin A/chemistry , Cyclosporine/pharmacology , Hepacivirus/physiology , Virus Replication/drug effects , Cell Line , Dose-Response Relationship, Drug , Humans , Isomerism
...