Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Ann Hematol ; 101(3): 557-569, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34981142

ABSTRACT

CC-90002 is an anti-CD47 antibody that inhibits CD47-SIRPα interaction and enables macrophage-mediated killing of tumor cells in hematological cancer cell lines. In this first clinical, phase 1, dose-escalation and -expansion study (CC-90002-AML-001; NCT02641002), we evaluated CC-90002 in patients with relapsed/refractory acute myeloid leukemia (AML) or high-risk myelodysplastic syndromes (MDS). CC-90002 was administered in escalating doses of 0.1-4.0 mg/kg, using a modified 3 + 3 design. Primary endpoints included dose-limiting toxicities (DLTs), non-tolerated dose (NTD), maximum tolerated dose (MTD), and recommended phase 2 dose. Secondary endpoints included preliminary efficacy, pharmacokinetics, and presence/frequency of anti-drug antibodies (ADAs). Between March 2016 and July 2018, 28 patients were enrolled (24 with AML and 4 with MDS) at 6 sites across the USA. As of July 18, 2018, all patients had discontinued, mainly due to death or progressive disease. The most common treatment-emergent adverse events were diarrhea (46.4%), thrombocytopenia (39.3%), febrile neutropenia (35.7%), and aspartate aminotransferase increase (35.7%). Four patients experienced DLTs (1 patient had grade 4 disseminated intravascular coagulation and grade 5 cerebral hemorrhage, 1 had grade 3 purpura, 1 had grade 4 congestive cardiac failure and grade 5 acute respiratory failure, and another had grade 5 sepsis). The NTD and MTD were not reached. No objective responses occurred. CC-90002 serum exposure was dose-dependent. ADAs were present across all doses, and the proportion of ADA-positive patients in cycle 1 increased over time. Despite no unexpected safety findings, the CC-90002-AML-001 study was discontinued in dose escalation for lack of monotherapy activity and evidence of ADAs. However, as other anti-CD47 agents in clinical trials are showing promising early results for AML and MDS, understanding preclinical and clinical differences between individual agents in this class will be of high importance.


Subject(s)
Antineoplastic Agents, Immunological , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Neoplasm Recurrence, Local , Adult , Aged , Aged, 80 and over , Animals , Female , Humans , Male , Mice , Middle Aged , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , CD47 Antigen/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Macaca fascicularis , Maximum Tolerated Dose , Mice, SCID , Myelodysplastic Syndromes/drug therapy , Neoplasm Recurrence, Local/drug therapy
2.
Cancer Immunol Immunother ; 71(2): 473-489, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34247273

ABSTRACT

Cluster of differentiation 47 (CD47) is a transmembrane protein ubiquitously expressed on human cells but overexpressed on many different tumor cells. The interaction of CD47 with signal-regulatory protein alpha (SIRPα) triggers a "don't eat me" signal to the macrophage, inhibiting phagocytosis. Thus, overexpression of CD47 enables tumor cells to escape from immune surveillance via the blockade of phagocytic mechanisms. We report here the development and characterization of CC-90002, a humanized anti-CD47 antibody. CC-90002 is unique among previously reported anti-CD47 bivalent antibodies that it does not promote hemagglutination while maintaining high-affinity binding to CD47 and inhibition of the CD47-SIRPα interaction. Studies in a panel of hematological cancer cell lines showed concentration-dependent CC-90002-mediated phagocytosis in acute lymphoblastic leukemia, acute myeloid leukemia (AML), lenalidomide-resistant multiple myeloma (MM) cell lines and AML cells from patients. In vivo studies with MM cell line-derived xenograft models established in immunodeficient mice demonstrated significant dose-dependent antitumor activity of CC-90002. Treatment with CC-90002 significantly prolonged survival in an HL-60-disseminated AML model. Mechanistic studies confirmed the binding of CC-90002 to tumor cells and concomitant recruitment of F4-80 positive macrophages into the tumor and an increase in expression of select chemokines and cytokines of murine origin. Furthermore, the role of macrophages in the CC-90002-mediated antitumor activity was demonstrated by transient depletion of macrophages with liposome-clodronate treatment. In non-human primates, CC-90002 displayed acceptable pharmacokinetic properties and a favorable toxicity profile. These data demonstrate the potential activity of CC-90002 across hematological malignancies and provided basis for clinical studies CC-90002-ST-001 (NCT02367196) and CC-90002-AML-001 (NCT02641002).


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, Differentiation/immunology , CD47 Antigen/immunology , Immunoglobulin Fc Fragments/immunology , Leukemia, Promyelocytic, Acute/drug therapy , Macrophages/immunology , Receptors, Immunologic/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, Differentiation/metabolism , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacology , Apoptosis , CD47 Antigen/metabolism , Cell Differentiation , Cell Proliferation , Female , Humans , Leukemia, Promyelocytic, Acute/immunology , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Phagocytosis , Prognosis , Receptors, Immunologic/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Cancer Manag Res ; 11: 10463-10476, 2019.
Article in English | MEDLINE | ID: mdl-31853198

ABSTRACT

PURPOSE: This first-in-human Phase I study investigated the safety, pharmacokinetics (PK), pharmacodynamic profile, and preliminary efficacy of CC-115, a dual inhibitor of mammalian target of rapamycin (mTOR) kinase and DNA-dependent protein kinase. PATIENTS AND METHODS: Patients with advanced solid or hematologic malignancies were enrolled in dose-finding and cohort expansion phases. In dose-finding, once-daily or twice-daily (BID) ascending oral doses of CC-115 (range: 0.5-40 mg/day) in 28-day continuous cycles identified the maximum-tolerated dose for cohort expansion in 5 specified tumor types. Twelve additional patients with mixed solid tumors participated in a bioavailability substudy. RESULTS: Forty-four patients were enrolled in the dose-finding cohort. Dose-limiting toxicity included thrombocytopenia, stomatitis, hyperglycemia, asthenia/fatigue, and increased transaminases. CC-115 10 mg BID was selected for cohort expansion (n=74) in which fatigue, nausea, and decreased appetite were the most frequent toxicities. Dose-proportional PK was found. CC-115 distributed to glioblastoma tissue (mean tumor/plasma concentration ratio: 0.713). Total exposure of CC-115 was similar under fasting and fed conditions. A patient with endometrial carcinoma remained in complete remission >4 years. Partial response (PR; n=2) and stable disease (SD; n=4) were reported in the bioavailability substudy; SD was reached in 53%, 22%, 21%, and 64% of patients with head and neck squamous cell carcinoma, Ewing sarcoma, glioblastoma multiforme, and castration-resistant prostate cancer, respectively. Chronic lymphocytic leukemia/small lymphocytic lymphoma showed 38% PR and 25% SD. CONCLUSION: CC-115 was well-tolerated, with toxicities consistent with mTOR inhibitors. Together with biomarker inhibition and preliminary efficacy, oral CC-115 10 mg BID is a promising novel anticancer treatment. CLINICAL TRIAL REGISTRATION: NCT01353625.

4.
Clin Cancer Res ; 25(18): 5623-5637, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31266833

ABSTRACT

PURPOSE: DNA-dependent protein kinase catalytic subunit (DNA-PK) is a pleiotropic kinase involved in DNA repair and transcriptional regulation. DNA-PK is deregulated in selected cancer types and is strongly associated with poor outcome. The underlying mechanisms by which DNA-PK promotes aggressive tumor phenotypes are not well understood. Here, unbiased molecular investigation in clinically relevant tumor models reveals novel functions of DNA-PK in cancer.Experimental Design: DNA-PK function was modulated using both genetic and pharmacologic methods in a series of in vitro models, in vivo xenografts, and patient-derived explants (PDE), and the impact on the downstream signaling and cellular cancer phenotypes was discerned. Data obtained were used to develop novel strategies for combinatorial targeting of DNA-PK and hormone signaling pathways. RESULTS: Key findings reveal that (i) DNA-PK regulates tumor cell proliferation; (ii) pharmacologic targeting of DNA-PK suppresses tumor growth both in vitro, in vivo, and ex vivo; (iii) DNA-PK transcriptionally regulates the known DNA-PK-mediated functions as well as novel cancer-related pathways that promote tumor growth; (iv) dual targeting of DNA-PK/TOR kinase (TORK) transcriptionally upregulates androgen signaling, which can be mitigated using the androgen receptor (AR) antagonist enzalutamide; (v) cotargeting AR and DNA-PK/TORK leads to the expansion of antitumor effects, uncovering the modulation of novel, highly relevant protumorigenic cancer pathways; and (viii) cotargeting DNA-PK/TORK and AR has cooperative growth inhibitory effects in vitro and in vivo. CONCLUSIONS: These findings uncovered novel DNA-PK transcriptional regulatory functions and led to the development of a combinatorial therapeutic strategy for patients with advanced prostate cancer, currently being tested in the clinical setting.


Subject(s)
DNA-Activated Protein Kinase/metabolism , Neoplasms/metabolism , Androgen Receptor Antagonists/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptors, Androgen/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription, Genetic , Xenograft Model Antitumor Assays
5.
Mol Cancer Ther ; 17(8): 1727-1738, 2018 08.
Article in English | MEDLINE | ID: mdl-29866747

ABSTRACT

Historically, phenotypic-based drug discovery has yielded a high percentage of novel drugs while uncovering new tumor biology. CC-671 was discovered using a phenotypic screen for compounds that preferentially induced apoptosis in triple-negative breast cancer cell lines while sparing luminal breast cancer cell lines. Detailed in vitro kinase profiling shows CC-671 potently and selectively inhibits two kinases-TTK and CLK2. Cellular mechanism of action studies demonstrate that CC-671 potently inhibits the phosphorylation of KNL1 and SRp75, direct TTK and CLK2 substrates, respectively. Furthermore, CC-671 causes mitotic acceleration and modification of pre-mRNA splicing leading to apoptosis, consistent with cellular TTK and CLK inhibition. Correlative analysis of genomic and potency data against a large panel of breast cancer cell lines identifies breast cancer cells with a dysfunctional G1-S checkpoint as more sensitive to CC-671, suggesting synthetic lethality between G1-S checkpoint and TTK/CLK2 inhibition. Furthermore, significant in vivo CC-671 efficacy was demonstrated in two cell line-derived and one patient tumor-derived xenograft models of triple-negative breast cancer (TNBC) following weekly dosing. These findings are the first to demonstrate the unique inhibitory combination activity of a dual TTK/CLK2 inhibitor that preferably kills TNBC cells and shows synthetic lethality with a compromised G1-S checkpoint in breast cancer cell lines. On the basis of these data, CC-671 was moved forward for clinical development as a potent and selective TTK/CLK2 inhibitor in a subset of patients with TNBC. Mol Cancer Ther; 17(8); 1727-38. ©2018 AACR.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Synthetic Lethal Mutations/drug effects , Animals , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Female , Humans , Mice , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Triple Negative Breast Neoplasms/drug therapy
6.
Oncotarget ; 8(43): 74688-74702, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-29088817

ABSTRACT

CC-115, a selective dual inhibitor of the mammalian target of rapamycin (mTOR) kinase and DNA-dependent protein kinase (DNA-PK), is undergoing Phase 1 clinical studies. Here we report the characterization of DNA-PK inhibitory activity of CC-115 in cancer cell lines. CC-115 inhibits auto-phosphorylation of the catalytic subunit of DNA-PK (DNA-PKcs) at the S2056 site (pDNA-PK S2056), leading to blockade of DNA-PK-mediated non-homologous end joining (NHEJ). CC-115 also indirectly reduces the phosphorylation of ataxia-telangiectasia mutated kinase (ATM) at S1981 and its substrates as well as homologous recombination (HR). The mTOR kinase and DNA-PK inhibitory activity of CC-115 leads to not only potent anti-tumor activity against a large panel of hematopoietic and solid cancer cell lines but also strong induction of apoptosis in a subset of cancer lines. Mechanistically, CC-115 prevents NHEJ by inhibiting the dissociation of DNA-PKcs, X-ray repair cross-complementing protein 4 (XRCC4), and DNA ligase IV from DNA ends. CC-115 inhibits colony formation of ATM-deficient cells more potently than ATM-proficient cells, indicating that inhibition of DNA-PK is synthetically lethal with the loss of functional ATM. In conclusion, CC-115 inhibits both mTOR signaling and NHEJ and HR by direct inhibition of DNA-PK. The mechanistic data not only provide selection of potential pharmacodynamic (PD) markers but also support CC-115 clinical development in patients with ATM-deficient tumors.

7.
Br J Haematol ; 172(6): 889-901, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26914976

ABSTRACT

Pomalidomide is an IMiD(®) immunomodulatory agent, which has shown clinically significant benefits in relapsed and/or refractory multiple myeloma (rrMM) patients when combined with dexamethasone, regardless of refractory status to lenalidomide or bortezomib. (Schey et al, ; San Miguel et al, 2013; Richardson et al, 2014; Scott, ) In this work, we present preclinical data showing that the combination of pomalidomide with dexamethasone (PomDex) demonstrates potent anti-proliferative and pro-apoptotic activity in both lenalidomide-sensitive and lenalidomide-resistant MM cell lines. PomDex also synergistically inhibited tumour growth compared with single-agent treatment in xenografts of lenalidomide-resistant H929 R10-1 cells. Typical hallmarks of IMiD compound activity, including IKZF3 (Aiolos) degradation, and the downregulation of interferon regulatory factor (IRF) 4 and MYC, seen in lenalidomide-sensitive H929 MM cell lines, were also observed in PomDex-treated lenalidomide-resistant H929 MM cells. Remarkably, this resulted in strong, synergistic effects on the induction of apoptosis in both lenalidomide-sensitive and resistant MM cells. Furthermore, gene expression profiling revealed a unique differential gene expression pattern in PomDex-treated samples, highlighted by the modulation of pro-apoptotic pathways in lenalidomide-resistant cells. These results provide key insights into molecular mechanisms of PomDex in the lenalidomide-resistant setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Dexamethasone/administration & dosage , Drug Resistance, Neoplasm , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunomodulation/drug effects , Lenalidomide , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
8.
J Med Chem ; 58(13): 5323-33, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26083478

ABSTRACT

We report here the synthesis and structure-activity relationship (SAR) of a novel series of mammalian target of rapamycin (mTOR) kinase inhibitors. A series of 4,6- or 1,7-disubstituted-3,4-dihydropyrazino[2,3-b]pyrazine-2(1H)-ones were optimized for in vivo efficacy. These efforts resulted in the identification of compounds with excellent mTOR kinase inhibitory potency, with exquisite kinase selectivity over the related lipid kinase PI3K. The improved PK properties of this series allowed for exploration of in vivo efficacy and ultimately the selection of CC-223 for clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Humans , Male , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Pyrazines/chemical synthesis , Rats , Structure-Activity Relationship , Tumor Cells, Cultured
9.
J Med Chem ; 58(14): 5599-608, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26102506

ABSTRACT

We report here the synthesis and structure-activity relationship (SAR) of a novel series of triazole containing mammalian target of rapamycin (mTOR) kinase inhibitors. SAR studies examining the potency, selectivity, and PK parameters for a series of triazole containing 4,6- or 1,7-disubstituted-3,4-dihydropyrazino[2,3-b]pyrazine-2(1H)-ones resulted in the identification of triazole containing mTOR kinase inhibitors with improved PK properties. Potent compounds from this series were found to block both mTORC1(pS6) and mTORC2(pAktS473) signaling in PC-3 cancer cells, in vitro and in vivo. When assessed in efficacy models, analogs exhibited dose-dependent efficacy in tumor xenograft models. This work resulted in the selection of CC-115 for clinical development.


Subject(s)
Drug Design , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazines/chemistry , Pyrazines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triazoles/chemistry , Triazoles/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Protein Conformation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Pyrazines/metabolism , Pyrazines/pharmacokinetics , Rats , Signal Transduction/drug effects , Structure-Activity Relationship , TOR Serine-Threonine Kinases/chemistry , TOR Serine-Threonine Kinases/metabolism , Triazoles/metabolism , Triazoles/pharmacokinetics , Xenograft Model Antitumor Assays
10.
Blood ; 126(6): 779-89, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26002965

ABSTRACT

Cereblon (CRBN), a substrate receptor of the Cullin 4 RING E3 ubiquitin ligase complex, is the target of the immunomodulatory drugs lenalidomide and pomalidomide. Recently, it was demonstrated that binding of these drugs to CRBN promotes the ubiquitination and subsequent degradation of 2 common substrates, transcription factors Aiolos and Ikaros. Here we report that CC-122, a new chemical entity termed pleiotropic pathway modifier, binds CRBN and promotes degradation of Aiolos and Ikaros in diffuse large B-cell lymphoma (DLBCL) and T cells in vitro, in vivo, and in patients, resulting in both cell autonomous as well as immunostimulatory effects. In DLBCL cell lines, CC-122-induced degradation or short hairpin RNA-mediated knockdown of Aiolos and Ikaros correlates with increased transcription of interferon (IFN)-stimulated genes independent of IFN-α, -ß, and -γ production and/or secretion and results in apoptosis in both activated B-cell (ABC) and germinal center B-cell DLBCL cell lines. Our results provide mechanistic insight into the cell-of-origin independent antilymphoma activity of CC-122, in contrast to the ABC subtype selective activity of lenalidomide.


Subject(s)
Antineoplastic Agents/pharmacology , B-Lymphocytes/drug effects , Ikaros Transcription Factor/genetics , Lymphoma, Large B-Cell, Diffuse/drug therapy , Peptide Hydrolases/genetics , Piperidones/pharmacology , Quinazolinones/pharmacology , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing , Animals , Antineoplastic Agents/chemistry , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Ikaros Transcription Factor/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Interferons/genetics , Interferons/metabolism , Lenalidomide , Lentivirus/genetics , Lentivirus/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, SCID , Molecular Mimicry , Peptide Hydrolases/metabolism , Piperidones/chemistry , Proteolysis/drug effects , Quinazolinones/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Ubiquitin-Protein Ligases , Xenograft Model Antitumor Assays
11.
Mol Cancer Ther ; 14(6): 1295-305, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25855786

ABSTRACT

mTOR is a serine/threonine kinase that regulates cell growth, metabolism, proliferation, and survival. mTOR complex-1 (mTORC1) and mTOR complex-2 (mTORC2) are critical mediators of the PI3K-AKT pathway, which is frequently mutated in many cancers, leading to hyperactivation of mTOR signaling. Although rapamycin analogues, allosteric inhibitors that target only the mTORC1 complex, have shown some clinical activity, it is hypothesized that mTOR kinase inhibitors, blocking both mTORC1 and mTORC2 signaling, will have expanded therapeutic potential. Here, we describe the preclinical characterization of CC-223. CC-223 is a potent, selective, and orally bioavailable inhibitor of mTOR kinase, demonstrating inhibition of mTORC1 (pS6RP and p4EBP1) and mTORC2 [pAKT(S473)] in cellular systems. Growth inhibitory activity was demonstrated in hematologic and solid tumor cell lines. mTOR kinase inhibition in cells, by CC-223, resulted in more complete inhibition of the mTOR pathway biomarkers and improved antiproliferative activity as compared with rapamycin. Growth inhibitory activity and apoptosis was demonstrated in a panel of hematologic cancer cell lines. Correlative analysis revealed that IRF4 expression level associates with resistance, whereas mTOR pathway activation seems to associate with sensitivity. Treatment with CC-223 afforded in vivo tumor biomarker inhibition in tumor-bearing mice, after a single oral dose. CC-223 exhibited dose-dependent tumor growth inhibition in multiple solid tumor xenografts. Significant inhibition of mTOR pathway markers pS6RP and pAKT in CC-223-treated tumors suggests that the observed antitumor activity of CC-223 was mediated through inhibition of both mTORC1 and mTORC2. CC-223 is currently in phase I clinical trials.


Subject(s)
Neoplasms/drug therapy , Pyrazines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , HCT116 Cells , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice, SCID , Molecular Structure , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/metabolism , Neoplasms/blood supply , Neoplasms/metabolism , Neovascularization, Pathologic/prevention & control , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazines/chemistry , TOR Serine-Threonine Kinases/metabolism , Tumor Burden/drug effects
12.
Br J Haematol ; 165(6): 870-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24635723

ABSTRACT

Sotatercept (ACE-011), a recombinant human fusion protein containing the extracellular domain of the human Activin receptor IIA, binds to and inhibits activin and other members of the transforming growth factor -ß (TGF-ß) superfamily. Administration of sotatercept led to a rapid and sustained increase in red blood cell (RBC) count and haemoglobin (Hb) in healthy volunteers (phase I clinical trials), but the mechanism is not fully understood. Mice treated with RAP-011 (murine ortholog of ACE-011) respond with a rapid (within 24 h) increase in haematocrit, Hb, and RBC count. These effects are accompanied by an equally rapid stimulation of late-stage erythroid precursors in the bone marrow (BM). RAP-011 also induces a significant increase in erythroid burst-forming units and erythropoietin, which could contribute to additional, sustained effects on RBC production. Further in vitro co-culture studies demonstrate that BM accessory cells are required for RAP-011 effects. To better understand which TGF-ß family ligand(s) mediate RAP-011 effects, we evaluated the impact of several of these ligands on erythroid differentiation. Our data suggest that RAP-011 may act to rescue growth differentiation factor 11/Activin A-induced inhibition of late-stage erythropoiesis. These data define the mechanism of action of a novel agent that regulates RBC differentiation and provide the rationale to develop sotatercept for the treatment of anaemia and ineffective erythropoiesis.


Subject(s)
Activin Receptors, Type II/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythropoiesis/drug effects , Erythropoiesis/physiology , Hemoglobins/biosynthesis , Recombinant Fusion Proteins/pharmacology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cellular Microenvironment/physiology , Colony-Forming Units Assay , Erythrocyte Indices/drug effects , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoietin/biosynthesis , Female , Humans , Ligands , Mice , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism
13.
Clin Cancer Res ; 19(20): 5722-32, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24030701

ABSTRACT

PURPOSE: mTOR pathway hyperactivation occurs in approximately 90% of glioblastomas, but the allosteric mTOR inhibitor rapamycin has failed in the clinic. Here, we examine the efficacy of the newly discovered ATP-competitive mTOR kinase inhibitors CC214-1 and CC214-2 in glioblastoma, identifying molecular determinants of response and mechanisms of resistance, and develop a pharmacologic strategy to overcome it. EXPERIMENTAL DESIGN: We conducted in vitro and in vivo studies in glioblastoma cell lines and an intracranial model to: determine the potential efficacy of the recently reported mTOR kinase inhibitors CC214-1 (in vitro use) and CC214-2 (in vivo use) at inhibiting rapamycin-resistant signaling and blocking glioblastoma growth and a novel single-cell technology-DNA Encoded Antibody Libraries-was used to identify mechanisms of resistance. RESULTS: Here, we show that CC214-1 and CC214-2 suppress rapamycin-resistant mTORC1 signaling, block mTORC2 signaling, and significantly inhibit the growth of glioblastomas in vitro and in vivo. EGFRvIII expression and PTEN loss enhance sensitivity to CC214 compounds, consistent with enhanced efficacy in strongly mTOR-activated tumors. Importantly, CC214 compounds potently induce autophagy, preventing tumor cell death. Genetic or pharmacologic inhibition of autophagy greatly sensitizes glioblastoma cells and orthotopic xenografts to CC214-1- and CC214-2-induced cell death. CONCLUSIONS: These results identify CC214-1 and CC214-2 as potentially efficacious mTOR kinase inhibitors in glioblastoma, and suggest a strategy for identifying patients most likely to benefit from mTOR inhibition. In addition, this study also shows a central role for autophagy in preventing mTOR-kinase inhibitor-mediated tumor cell death, and suggests a pharmacologic strategy for overcoming it.


Subject(s)
ErbB Receptors/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Imidazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Glioblastoma/drug therapy , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/metabolism , PTEN Phosphohydrolase/metabolism , Protein Biosynthesis/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
Bioorg Med Chem Lett ; 22(3): 1427-32, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22226655

ABSTRACT

In this Letter we describe the optimization of an aminopurine lead (1) with modest potency and poor overall kinase selectivity which led to the identification of a series of potent, selective JNK inhibitors. Improvement in kinase selectivity was enabled by introduction of an aliphatic side chain at the C-2 position. CC-359 (2) was selected as a potential clinical candidate for diseases manifested by ischemia reperfusion injury.


Subject(s)
2-Aminopurine/chemistry , 2-Aminopurine/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Purines/chemistry , Reperfusion Injury/enzymology , Animals , Catalytic Domain , Dogs , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Haplorhini , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Purines/pharmacology , Rats , Reperfusion Injury/drug therapy , Structure-Activity Relationship
15.
Bioorg Med Chem Lett ; 22(3): 1433-8, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22244937

ABSTRACT

In this Letter we describe the discovery of potent, selective, and orally active aminopurine JNK inhibitors. Improving the physico-chemical properties as well as increasing the potency and selectivity of a subseries with rat plasma exposure, led to the identification of four structurally diverse inhibitors. Differentiation based on PK profiles in multiple species as well as activity in a chronic efficacy model led to the identification of 1 (CC-930) as a development candidate, which is currently in Phase II clinical trial for IPF.


Subject(s)
Cyclohexanols/chemistry , Cyclohexanols/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , MAP Kinase Kinase 4/antagonists & inhibitors , Purines/chemistry , Purines/pharmacology , Administration, Oral , Animals , Catalytic Domain , Cyclohexanols/administration & dosage , Dogs , Enzyme Activation/drug effects , Enzyme Inhibitors/administration & dosage , Haplorhini , Idiopathic Pulmonary Fibrosis/drug therapy , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Purines/administration & dosage , Rats , Structure-Activity Relationship
16.
Bioorg Med Chem ; 19(21): 6356-74, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21955454

ABSTRACT

A series of 1,1-diarylalkene derivatives were prepared to optimize the properties of CC-5079 (1), a dual inhibitor of tubulin polymerization and phosphodiesterase 4 (PDE4). By using the 3-ethoxy-4-methoxyphenyl PDE4 pharmacophore as one of the aromatic rings, a significant improvement in PDE4 inhibition was achieved. Compound 28 was identified as a dual inhibitor with potent PDE4 (IC(50)=54 nM) and antitubulin activity (HCT-116 IC(50)=34 nM and tubulin polymerization IC(50) ∼1 µM). While the nitrile group at the alkene terminus was generally required for potent antiproliferative activity, its replacement was tolerated if there was a hydroxyl or amino group on one of the aryl rings. Conveniently, this group could also serve as a handle for amino acid derivatization to improve the compounds' solubility. The glycinamide analog 45 showed significant efficacy in the HCT-116 xenograft model, with 64% inhibition of tumor growth upon dosing at 20 mg/kg qd.


Subject(s)
Alkenes/chemistry , Alkenes/pharmacology , Antineoplastic Agents/chemistry , Benzene Derivatives/chemistry , Benzene Derivatives/pharmacology , Colorectal Neoplasms/drug therapy , Phosphodiesterase Inhibitors/chemistry , Tubulin Modulators/chemistry , Alkenes/chemical synthesis , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Benzene Derivatives/chemical synthesis , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , HCT116 Cells , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Molecular Dynamics Simulation , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/pharmacology , Structure-Activity Relationship , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/pharmacology
17.
J Med Chem ; 52(6): 1522-4, 2009 Mar 26.
Article in English | MEDLINE | ID: mdl-19256507

ABSTRACT

In this communication, we report the discovery of 1S (apremilast), a novel potent and orally active phosphodiesterase 4 (PDE4) and tumor necrosis factor-alpha inhibitor. The optimization of previously reported 3-(1,3-dioxo-1,3-dihydroisoindol-2-yl)-3-(3,4-dimethoxyphenyl)propionic acid PDE4 inhibitors led to this series of sulfone analogues. Evaluation of the structure-activity relationship of substitutions on the phthalimide group led to the discovery of an acetylamino analogue 1S, which is currently in clinical trials.


Subject(s)
Drug Discovery , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Thalidomide/analogs & derivatives , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Administration, Oral , Animals , Humans , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/chemistry , Rats , Stereoisomerism , Structure-Activity Relationship , Thalidomide/administration & dosage , Thalidomide/chemistry , Thalidomide/pharmacology
18.
Mol Cell Neurosci ; 34(3): 390-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17222562

ABSTRACT

We have studied the stability of the dopaminergic phenotype in a conditionally immortalized human mesencephalic cell line, MESC2.10. Even though MESC2.10 cells exhibit features of dopaminergic neurons in vitro, none of the cells expressed tyrosine hydroxylase (TH) after transplantation into a rat model of Parkinson's disease. We examined whether this is caused by cell death or loss of transmitter phenotype. Cells were cultured in differentiation medium, then harvested and replated into the same medium where they continued to express TH, whereas replated cells fed medium lacking differentiation factors (dibutyryl cAMP and glial cell line-derived neurotrophic factor) did not. Interestingly, cultures grown in the absence of differentiation factors could regain TH expression once exposed to differentiation medium. Our data suggest that TH expression in vitro is inducible in neurons derived from the MESC2.10 cell line and that the dopaminergic phenotype of these cells in vivo might be unstable.


Subject(s)
Brain Tissue Transplantation , Gene Expression/physiology , Mesencephalon/cytology , Neurons/enzymology , Tyrosine 3-Monooxygenase/metabolism , Analysis of Variance , Animals , Behavior, Animal , Cell Differentiation/drug effects , Cell Line , Cyclic AMP/pharmacology , Disease Models, Animal , Female , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Humans , Indoles , Karyotyping/methods , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/surgery , Rats , Tubulin/metabolism
19.
Cancer Res ; 66(2): 951-9, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16424030

ABSTRACT

We have found that the synthetic compound CC-5079 potently inhibits cancer cell growth in vitro and in vivo by a novel combination of molecular mechanisms. CC-5079 inhibits proliferation of cancer cell lines from various organs and tissues at nanomolar concentrations. Its IC(50) value ranges from 4.1 to 50 nmol/L. The effect of CC-5079 on cell growth is associated with cell cycle arrest in G(2)-M phase, increased phosphorylation of G(2)-M checkpoint proteins, and apoptosis. CC-5079 prevents polymerization of purified tubulin in a concentration-dependent manner in vitro and depolymerizes microtubules in cultured cancer cells. In competitive binding assays, CC-5079 competes with [(3)H]colchicine for binding to tubulin; however, it does not compete with [(3)H]paclitaxel (Taxol) or [(3)H]vinblastine. Our data indicate that CC-5079 inhibits cancer cell growth with a mechanism of action similar to that of other tubulin inhibitors. However, CC-5079 remains active against multidrug-resistant cancer cells unlike other tubulin-interacting drugs, such as Taxol and colchicine. Interestingly, CC-5079 also inhibits tumor necrosis factor-alpha (TNF-alpha) secretion from lipopolysaccharide-stimulated human peripheral blood mononuclear cells (IC(50), 270 nmol/L). This inhibitory effect on TNF-alpha production is related to its inhibition of phosphodiesterase type 4 enzymatic activity. Moreover, in a mouse xenograft model using HCT-116 human colorectal tumor cells, CC-5079 significantly inhibits tumor growth in vivo. In conclusion, our data indicate that CC-5079 represents a new chemotype with novel mechanisms of action and that it has the potential to be developed for neoplastic and inflammatory disease therapy.


Subject(s)
Nitriles/pharmacology , Tubulin/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Neoplasms/pathology , Transplantation, Heterologous , Tubulin Modulators/pharmacology , Tumor Cells, Cultured
20.
Expert Opin Emerg Drugs ; 8(1): 71-81, 2003 May.
Article in English | MEDLINE | ID: mdl-14610913

ABSTRACT

Despite the availability of a great number of medications, the asthma epidemic is continuing to increase. It is obvious that a high, unmet medical need remains and innovative therapeutic agents are urgently required. Existing therapies, such as beta-agonists and corticosteroids, provide relief for sufferers of mild-to-moderate asthma, reversing the acute bronchoconstriction and decreasing the inflammation. However, these therapies provide little relief for chronic asthmatics. Asthma is a manifestation of an imbalance in cytokine and signalling pathways that mediate inflammatory and structural changes within the lung. New therapies need to be developed to target these changes. Emerging treatments for asthma include strategies to alter the cytokine/chemokine balance, to skew the cytokine profile away from a T helper (Th)2 response and towards a Th1 response. Strategies designed to do this include therapeutic antibodies or small molecule inhibitors targeted towards IL-13, IL-4 or their receptors, and the Th1 cytokine IL-12. Much interest has focused on the signalling pathways involved in asthma. Among these, the mitogen-activated protein kinase (MAPK) pathway members c-Jun N-terminal kinase (JNK) and p38 have gathered much interest, in addition to the transcription factors nuclear factor kappaB (NF-kappa B), activator protein-1 (AP-1) and signal transducer and activator of transcription (STAT)-6. This review aims to summarise the emerging treatments for chronic asthma, from early discovery, to late clinical stages, and discuss the therapeutic rationale behind these treatments. Much is still to be learned about the mechanisms involved in the development and treatment of chronic asthma; however, much promise lies in the future of these new therapeutics.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Drug Delivery Systems/trends , Animals , Anti-Asthmatic Agents/immunology , Asthma/immunology , Drug Delivery Systems/methods , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...