Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
Int J Mol Sci ; 23(23)2022 Nov 22.
Article in English | MEDLINE | ID: mdl-36498832

ABSTRACT

Epilepsy is one of the common encephalopathies caused by sudden abnormal discharges of neurons in the brain. About 30% of patients with epilepsy are insensitive and refractory to existing antiseizure medications. The sonic hedgehog signaling pathway is essential to the development and homeostasis of brain. Aberrant sonic hedgehog signaling is increased in refractory epileptic lesions and may involve the etiology of epilepsy. Thus, new inhibitors of Smoothened, a key signal transducer of this signaling pathway are urgently need for refractory epilepsy. We have established a high-throughput screening platform and discovered several active small molecules targeting Smoothened including TT22. Here we show that the novel Smoothened inhibitor TT22 could block the translocation of ßarrestin2-GFP to Smoothened, reduce the accumulation of Smoothened on primary cilia, displace Bodipy-cyclopamine binding to Smoothened, and inhibit the expression of downstream Gli transcription factor. Moreover, TT22 inhibits the abnormal seizure-like activity in neurons. Furthermore, we demonstrated that FDA-approved Smoothened inhibitor GDC-0449 and LDE-225 are able to inhibit abnormal seizure-like activity in neurons. Thus, our study suggests that targeting the sonic hedgehog signaling with new small-molecule Smoothened inhibitors might provide a potential new therapeutic avenue for refractory epilepsy.


Subject(s)
Drug Resistant Epilepsy , Hedgehog Proteins , Smoothened Receptor , Humans , Hedgehog Proteins/metabolism , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Seizures , Signal Transduction/physiology , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/metabolism
2.
Cells ; 11(20)2022 10 13.
Article in English | MEDLINE | ID: mdl-36291078

ABSTRACT

Basal cell carcinoma (BCC) is one of the most common neoplasms in the population. A good prognosis and mainly non-aggressive development have made it underdiagnosed and excluded from the statistics. Due to the availability of efficient surgical therapy, BCC is sometimes overlooked in the search for novel therapies. Most clinicians are unaware of its complicated pathogenesis or the availability of effective targeted therapy based on Hedgehog inhibitors (HHI) used in advanced or metastatic cases. Nevertheless, the concomitance and esthetic burden of this neoplasm are severe. As with other cancers, its pathogenesis is multifactorial and complicated with a network of dependencies. Although the tumour microenvironment (TME), genetic aberrations, and risk factors seem crucial in all skin cancers, in BCC they all have become accessible as therapeutic or prevention targets. The results of this review indicate that a central role in the development of BCC is played by the Hedgehog (Hh) signalling pathway. Two signalling molecules have been identified as the main culprits, namely Patched homologue 1 (PTCH1) and, less often, Smoothened homologue (SMO). Considering effective immunotherapy for other neoplastic growths being introduced, implementing immunotherapy in advanced BCC is pivotal and beneficial. Up to now, the US Food and Drug Administration (FDA) has approved two inhibitors of SMO for the treatment of advanced BCC. Sonidegib and vismodegib are registered based on their efficacy in clinical trials. However, despite this success, limitations might occur during the therapy, as some patients show resistance to these molecules. This review aims to summarize novel options of targeted therapies in BCC and debate the mechanisms and clinical implications of tumor resistance.


Subject(s)
Antineoplastic Agents , Carcinoma, Basal Cell , Hedgehog Proteins , Skin Neoplasms , Smoothened Receptor , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Basal Cell/drug therapy , Carcinoma, Basal Cell/metabolism , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Tumor Microenvironment , United States , Patched-1 Receptor/metabolism , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/metabolism
3.
J Med Chem ; 64(18): 13830-13840, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34492176

ABSTRACT

Class F G protein-coupled receptors are characterized by a large extracellular domain (ECD) in addition to the common transmembrane domain (TMD) with seven α-helixes. For smoothened receptor (SMO), structural studies revealed dissected ECD and TMD, and their integrated assemblies. However, distinct assemblies were reported under different circumstances. Using an unbiased approach based on four series of cross-conjugated bitopic ligands, we explore the relationship between the active status and receptor assembly. Different activity dependency on the linker length for these bitopic ligands corroborates the various occurrences of SMO assembly. These results reveal a rigid "near" assembly for active SMO, which is in contrast to previous results. Conversely, inactive SMO adopts a free ECD, which would be remotely captured at "far" assembly by cholesterol. Altogether, we propose a mechanism of cholesterol flow-caused SMO activation involving an erection of ECD from far to near assembly.


Subject(s)
Hydroxycholesterols/metabolism , Smoothened Receptor/metabolism , Anilides/chemical synthesis , Anilides/metabolism , Animals , Binding Sites , HEK293 Cells , Humans , Hydroxycholesterols/chemical synthesis , Ligands , Mice , NIH 3T3 Cells , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/metabolism , Protein Domains , Pyridines/chemical synthesis , Pyridines/metabolism , Smoothened Receptor/agonists , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/chemistry
4.
Front Immunol ; 12: 737245, 2021.
Article in English | MEDLINE | ID: mdl-34580585

ABSTRACT

Allergic asthma is a common inflammatory airway disease in which Th2 immune response and inflammation are thought to be triggered by inhalation of environmental allergens. Many studies using mouse models and human tissues and genome-wide association have indicated that Sonic Hedgehog (Shh) and the Hedgehog (Hh) signaling pathway are involved in allergic asthma and that Shh is upregulated in the lung on disease induction. We used a papain-induced mouse model of allergic airway inflammation to investigate the impact of systemic pharmacological inhibition of the Hh signal transduction molecule smoothened on allergic airway disease induction and severity. Smoothened-inhibitor treatment reduced the induction of Shh, IL-4, and IL-13 in the lung and decreased serum IgE, as well as the expression of Smo, Il4, Il13, and the mucin gene Muc5ac in lung tissue. Smoothened inhibitor treatment reduced cellular infiltration of eosinophils, mast cells, basophils, and CD4+ T-cells to the lung, and eosinophils and CD4+ T-cells in the bronchoalveolar lavage. In the mediastinal lymph nodes, smoothened inhibitor treatment reduced the number of CD4+ T-cells, and the cell surface expression of Th2 markers ST2 and IL-4rα and expression of Th2 cytokines. Thus, overall pharmacological smoothened inhibition attenuated T-cell infiltration to the lung and Th2 function and reduced disease severity and inflammation in the airway.


Subject(s)
Anti-Asthmatic Agents/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Asthma/drug therapy , Benzimidazoles/administration & dosage , Chemotaxis, Leukocyte/drug effects , Cytokines/metabolism , Lung/drug effects , Phenylurea Compounds/administration & dosage , Pneumonia/drug therapy , Smoothened Receptor/antagonists & inhibitors , Th2 Cells/drug effects , Animals , Asthma/immunology , Asthma/metabolism , Disease Models, Animal , Female , Injections, Intraperitoneal , Lung/immunology , Lung/metabolism , Male , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/metabolism , Signal Transduction , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
5.
Int J Mol Sci ; 22(16)2021 Aug 04.
Article in English | MEDLINE | ID: mdl-34445078

ABSTRACT

The Smoothened (SMO) receptor is the most druggable target in the Hedgehog (HH) pathway for anticancer compounds. However, SMO antagonists such as vismodegib rapidly develop drug resistance. In this study, new SMO antagonists having the versatile purine ring as a scaffold were designed, synthesised, and biologically tested to provide an insight to their mechanism of action. Compound 4s was the most active and the best inhibitor of cell growth and selectively cytotoxic to cancer cells. 4s induced cell cycle arrest, apoptosis, a reduction in colony formation and downregulation of PTCH and GLI1 expression. BODIPY-cyclopamine displacement assays confirmed 4s is a SMO antagonist. In vivo, 4s strongly inhibited tumour relapse and metastasis of melanoma cells in mice. In vitro, 4s was more efficient than vismodegib to induce apoptosis in human cancer cells and that might be attributed to its dual ability to function as a SMO antagonist and apoptosis inducer.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Purines/pharmacology , Smoothened Receptor/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , HT29 Cells , Hedgehog Proteins/metabolism , Humans , Mice, Inbred C57BL , Neoplasms/metabolism , Purines/chemistry , Purines/therapeutic use , Signal Transduction/drug effects , Smoothened Receptor/metabolism
6.
Hematology ; 26(1): 518-528, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34314648

ABSTRACT

OBJECTIVE: Hypomethylating agents (HMAs) have been reported to target the Sonic Hedgehog (Shh) signaling pathway in myelodysplastic syndrome (MDS). However, the synergistic inhibitory effect of Smo inhibitor jervine and its combination with decitabine in MUTZ-1 cell lines remains lacking. METHODS: We used a CCK-8 assay to detect the in-vitro proliferation rate of MUTZ-1 cell lines. Besides, the Annexin V-FITC/PI double staining flow cytometry was utilized to detect the apoptosis rate and cell cycle changes. The expression levels of mRNA were quantified by using qRT-PCR, and the western blot was employed to detect the expression of proteins. RESULTS: We found that the single-agent jervine or decitabine can significantly inhibit the proliferation rate of MUTZ-1 cell lines, and this inhibitory effect is time-dependent and concentration-dependent. The combined intervention of the jervine and decitabine can more significantly inhibit cell proliferation, induce cell apoptosis, and block the G1 phase of the cell cycle. The combined intervention of the two drugs significantly reduced Smo and G1i-1 mRNA expression in MUTZ-1 cells. Furthermore, after combining both of the drug treatments, the proteins levels of Smo, G1i-1, PI3K, p-AKT, Bcl2, and Cyclin Dl were significantly downregulated, and Caspase-3 is upregulated, indicating that jervine with its combination of decitabine might be effective for controlling the proliferation, apoptosis, and cell cycle. CONCLUSION: The Smo inhibitor jervine and its combination with decitabine have a synergistic effect on the proliferation, cell cycle, and apoptosis of MUTZ-1 cells, and its mechanism may be achieved by interfering with the Shh signaling pathway.


Subject(s)
Decitabine/pharmacology , Hedgehog Proteins/metabolism , Signal Transduction/drug effects , Smoothened Receptor/antagonists & inhibitors , Veratrum Alkaloids/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Regulation/drug effects , Humans , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
7.
Oncogene ; 40(22): 3799-3814, 2021 06.
Article in English | MEDLINE | ID: mdl-33958721

ABSTRACT

Despite the development of new targeted and immune therapies, the prognosis of metastatic melanoma remains bleak. Therefore, it is critical to better understand the mechanisms controlling advanced melanoma to develop more effective treatment regimens. Hedgehog/GLI (HH/GLI) signaling inhibitors targeting the central pathway transducer Smoothened (SMO) have shown to be clinical efficacious in skin cancer; however, several mechanisms of non-canonical HH/GLI pathway activation limit their efficacy. Here, we identify a novel SOX2-BRD4 transcriptional complex driving the expression of GLI1, the final effector of the HH/GLI pathway, providing a novel mechanism of non-canonical SMO-independent activation of HH/GLI signaling in melanoma. Consistently, we find a positive correlation between the expression of GLI1 and SOX2 in human melanoma samples and cell lines. Further, we show that combined targeting of canonical HH/GLI pathway with the SMO inhibitor MRT-92 and of the SOX2-BRD4 complex using a potent Proteolysis Targeted Chimeras (PROTACs)-derived BRD4 degrader (MZ1), yields a synergistic anti-proliferative effect in melanoma cells independently of their BRAF, NRAS, and NF1 mutational status, with complete abrogation of GLI1 expression. Combination of MRT-92 and MZ1 strongly potentiates the antitumor effect of either drug as single agents in an orthotopic melanoma model. Together, our data provide evidence of a novel mechanism of non-canonical activation of GLI1 by the SOX2-BRD4 transcriptional complex, and describe the efficacy of a new combinatorial treatment for a subset of melanomas with an active SOX2-BRD4-GLI1 axis.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Dipeptides/pharmacology , Guanidines/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Heterocyclic Compounds, 3-Ring/pharmacology , Melanoma/drug therapy , SOXB1 Transcription Factors/metabolism , Transcription Factors/antagonists & inhibitors , Zinc Finger Protein GLI1/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Dipeptides/administration & dosage , Drug Synergism , Female , Guanidines/administration & dosage , Hedgehog Proteins/metabolism , Heterocyclic Compounds, 3-Ring/administration & dosage , Humans , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , Molecular Targeted Therapy , Signal Transduction/drug effects , Smoothened Receptor/antagonists & inhibitors , Spheroids, Cellular , Xenograft Model Antitumor Assays
8.
Expert Opin Drug Saf ; 20(8): 877-882, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33888008

ABSTRACT

Introduction: Hedgehog inhibitors are an alternative treatment option for patients with advanced BCCs not eligible for standard therapies due to lack of efficacy, high recurrence risk, and high-rate morbidity. Sonidegib, an oral smoothened antagonist, has been approved for the treatment of adult patients with locally advanced basal cell carcinoma. Several studies and randomized controlled trials have been conducted in order to evaluate the efficacy, safety, and tolerability of this new molecule.Areas covered: The aim of this article is to provide a complete overview on the use of sonidegib for the treatment of advanced BCCs describing the efficacy, safety, and drug tolerability of this drug.Expert opinion: Sonidegib, with a different pharmacokinetics profile from that of the other SMO-inhibitor vismodegib, demonstrated to be an efficacious and well-tolerated treatment in patients with locally advanced BCC. Although several drug-related adverse events have already been described, different strategies should be taken into account to better manage this small molecule while avoiding treatment discontinuation. The use of sonidegib as neoadjuvant therapy or combined with other hedgehog pathway inhibitors targeting different sites and to date, only available for pre-clinical studies, should also be considered.


Subject(s)
Biphenyl Compounds/administration & dosage , Carcinoma, Basal Cell/drug therapy , Pyridines/administration & dosage , Skin Neoplasms/drug therapy , Anilides/administration & dosage , Anilides/adverse effects , Anilides/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacokinetics , Carcinoma, Basal Cell/pathology , Hedgehog Proteins/antagonists & inhibitors , Humans , Pyridines/adverse effects , Pyridines/pharmacokinetics , Randomized Controlled Trials as Topic , Skin Neoplasms/pathology , Smoothened Receptor/antagonists & inhibitors
9.
Bioorg Med Chem ; 39: 116166, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33910157

ABSTRACT

Constitutive activation of Hedgehog (Hh) pathway is intimately related with the occurrence and development of several malignancies, such as medulloblastoma (MB) and other tumors. Therefore, small molecular inhibitors of Hh pathway are urgently needed. In this study, three new steroidal alkaloids, ⊿5 (20R, 24R) 23-oxo-24-methylsolacongetidine, ⊿5 (20S, 24R) 23-oxo-24-methylsolacongetidine and veralinine 3-O-α-l-rhamnopyranosyl-(1 â†’ 2)-ß-D-glucopyranoside, together with six known alkaloids, 20-epi-verazine, verazine, protoverine 15-(l)-2'-methylbutyrate, jervine, veramarine and ß1-chaconine, were isolated and determined from Veratrum grandiflorum Loes. The dual-luciferase bioassay indicated that all compounds exhibited significant inhibitions of Hh pathway with IC50 values of 0.72-14.31 µM against Shh-LIGHT 2 cells. To determine whether these Hh pathway inhibitors act with the Smoothened (Smo) protein, which is an important oncoprotein and target for this pathway, BODIPY-cyclopamine (BC) competitive binding assay was preferentially performed. Compared with BC alone, all compounds obviously reduced the fluorescence intensities of BC binding with Smo in Smo-overexpression HEK293T cells through fluorescence microscope and flow cytometer. By directly interacting with Smo, it revealed that they were actually novel natural Smo inhibitors. Then, their anti-tumor effects were investigated against the human MB cell line DAOY, which is a typical pediatric brain tumor cells line with highly expressed Hh pathway. Interestingly, most of compounds had slight proliferation inhibitions on DAOY cells after treatment for 24 h same as vismodegib, while ß1-chaconine showed the strongest inhibitory effect on the growth of DAOY with IC50 value of 5.35 µM. In conclusion, our studies valuably provide several novel natural Smo inhibitors for potential targeting treatment of Hh-dependent tumors.


Subject(s)
Alkaloids/isolation & purification , Alkaloids/pharmacology , Cell Proliferation/drug effects , Medulloblastoma/pathology , Smoothened Receptor/antagonists & inhibitors , Steroids/chemistry , Veratrum/chemistry , Alkaloids/chemistry , Cell Line, Tumor , HEK293 Cells , Humans , Molecular Structure , Spectrum Analysis/methods
10.
Cancer Res ; 81(11): 3105-3120, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33853831

ABSTRACT

Hedgehog signaling is aberrantly activated in hematologic malignancies and solid tumors, and targeting it is a promising therapeutic strategy against these cancers. Resistance to clinically available hedgehog-targeted Smoothened inhibitor (SMOi) drugs has become a critical issue in hedgehog-driven cancer treatment. Our previous studies identified inhibition of BET and CDK7 as two epigenetic/transcriptional-targeted therapeutic strategies for overcoming SMOi resistance, providing a promising direction for anti-hedgehog drug development. To uncover additional strategies for inhibiting aberrant hedgehog activity, here we performed CRISPR-Cas9 screening with an single-guide RNA library targeting epigenetic and transcriptional modulators in hedgehog-driven medulloblastoma cells, combined with tumor dataset analyses. Structure specific recognition protein 1 (SSRP1), a subunit of facilitates chromatin transcription (FACT) complex, was identified as a hedgehog-induced essential oncogene and therapeutic target in hedgehog-driven cancer. The FACT inhibitor CBL0137, which has entered clinical trials for cancer, effectively suppressed in vitro and in vivo growth of multiple SMOi-responsive and SMOi-resistant hedgehog-driven cancer models. Mechanistically, CBL0137 exerted anti-hedgehog activity by targeting transcription of GLI1 and GLI2, which are core transcription factors of the hedgehog pathway. SSRP1 bound the promoter regions of GLI1 and GLI2, while CBL0137 treatment substantially disrupted these interactions. Moreover, CBL0137 synergized with BET or CDK7 inhibitors to antagonize aberrant hedgehog pathway and growth of hedgehog-driven cancer models. Taken together, these results identify FACT inhibition as a promising epigenetic/transcriptional-targeted therapeutic strategy for treating hedgehog-driven cancers and overcoming SMOi resistance. SIGNIFICANCE: This study identifies FACT inhibition as an anti-hedgehog therapeutic strategy for overcoming resistance to Smoothened inhibitors and provides preclinical support for initiating clinical trials of FACT-targeted drug CBL0137 against hedgehog-driven cancers.


Subject(s)
Carbazoles/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Hedgehog Proteins/metabolism , High Mobility Group Proteins/antagonists & inhibitors , Medulloblastoma/drug therapy , Smoothened Receptor/antagonists & inhibitors , Transcriptional Elongation Factors/antagonists & inhibitors , Animals , Apoptosis , Cell Proliferation , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Female , Humans , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Clin Lymphoma Myeloma Leuk ; 21(4): e415-e422, 2021 04.
Article in English | MEDLINE | ID: mdl-33547022

ABSTRACT

Advancements in the understanding of the pathogenesis of acute myeloid leukemia (AML) have led to the introduction and approval of a number of novel drugs in AML. Glasdegib, an oral hedgehog pathway inhibitor, was approved in 2018 in combination with low-dose cytarabine for the treatment of newly diagnosed AML in patients unfit for intensive chemotherapy. In this review, we discuss the preclinical rationale for glasdegib, important clinical trials that led to glasdegib's approval, and future trials of glasdegib in AML and other myeloid diseases. Notably, 2 large randomized, placebo-controlled phase 3 trials (AML BRIGHT 1019) are currently recruiting patients with newly diagnosed AML to evaluate glasdegib in combination with intensive chemotherapy or azacitidine, depending on the patient's ability to tolerate induction chemotherapy. While glasdegib and low-dose cytarabine have been eclipsed by venetoclax and hypomethylating agent combinations for newly diagnosed AML in the United States, we discuss other areas where glasdegib may still have an opportunity to improve outcomes in this devastating disease.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzimidazoles/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Phenylurea Compounds/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azacitidine/therapeutic use , Benzimidazoles/pharmacology , Cell Line, Tumor , Clinical Trials as Topic , Cytarabine/therapeutic use , Drug Approval , Drug Evaluation, Preclinical , Humans , Induction Chemotherapy/adverse effects , Induction Chemotherapy/methods , Mice , Phenylurea Compounds/pharmacology , Signal Transduction/drug effects , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/metabolism , United States
12.
Clin Pharmacol Drug Dev ; 10(3): 272-282, 2021 03.
Article in English | MEDLINE | ID: mdl-32790066

ABSTRACT

Glasdegib is a potent, selective oral inhibitor of the Hedgehog signaling pathway. This phase 1 double-blind thorough QT study (NCT03162900) evaluated the effects of glasdegib on QTc interval. The study enrolled 36 healthy volunteers to receive a single dose of 150 mg glasdegib (representing a therapeutic dose), 300 mg glasdegib (representing a supratherapeutic dose), 400 mg moxifloxacin (positive control), or placebo under fasted conditions. The study demonstrated that therapeutic and supratherapeutic doses of glasdegib had no significant effect on QTc interval; the upper bound of the 2-sided 90% confidence intervals (CIs) for all time-matched least-squares mean differences in QT interval corrected using Fridericia's formula (QTcF) between glasdegib and placebo was below the prespecified criterion of 20 milliseconds (Food and Drug Administration correspondence reviewed and accepted). Based on an exposure-response analysis, glasdegib was determined not to have a meaningful effect on heart rate (change in RR interval). The mean (90%CI) model-derived baseline and placebo-adjusted QTcF at the average maximum observed concentration values corresponding to therapeutic and supratherapeutic glasdegib doses was 7.3 milliseconds (6.5-8.2 milliseconds) and 13.7 milliseconds (12.0-15.5 milliseconds), respectively. Together these results demonstrated that following therapeutic and supratherapeutic glasdegib dosing, the change in QTc from baseline was well below the 20-millisecond threshold of clinical concern in oncology.


Subject(s)
Benzimidazoles/pharmacokinetics , Heart/drug effects , Hedgehog Proteins/antagonists & inhibitors , Phenylurea Compounds/pharmacokinetics , Smoothened Receptor/antagonists & inhibitors , Adult , Benzimidazoles/pharmacology , Case-Control Studies , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Electrocardiography/drug effects , Electrocardiography/methods , Fasting , Healthy Volunteers/statistics & numerical data , Heart/physiology , Humans , Male , Middle Aged , Moxifloxacin/administration & dosage , Phenylurea Compounds/pharmacology , Placebos/administration & dosage , Topoisomerase II Inhibitors/administration & dosage
13.
Acta Pharmacol Sin ; 42(6): 1005-1013, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32855528

ABSTRACT

Aberrantly activated Hedgehog (Hh) pathway is critical for driving the initiation and progression of multiple types of cancers, including medulloblastoma (MB) and basal cellular carcinoma (BCC). The majority of current Hh antagonist function by targeting the transmembrane domain of the oncoprotein Smoothened (Smo), a G-protein-coupled receptor-like receptor of Hh pathway. However, the primary and acquired resistance to current Smo inhibitors raise a critical need to develop next-generation of Smo inhibitors to improve their clinical efficacy. In this study, we identify that FDA approved drug ABT-199 significantly and selectively inhibits the Hh pathway. Mechanistically, ABT-199 acts as a competitive inhibitor of oxysterol by potentially targeting the cysteine rich domain (CRD) of Smo, rather as a BH3 mimetic. ABT-199 obviously inhibits the growth of Hh-driven tumors and possesses capacity of combating the primary and acquired resistance to Smo inhibitors caused by Smo mutations. Our data reposition ABT-199 as a Smo inhibitor for treating Hh-driven tumors, especially for those bearing Smo mutations and resistant to current Smo inhibitors. Meanwhile, our findings strengthen the argument that the CRD of Smo is a promising target for developing novel Smo inhibitors with capacity of combating the resistance to Smo inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Neoplasms/drug therapy , Signal Transduction/drug effects , Smoothened Receptor/antagonists & inhibitors , Sulfonamides/therapeutic use , Animals , Antineoplastic Agents/metabolism , Binding Sites , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Hedgehog Proteins/metabolism , Humans , Hydroxycholesterols/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , NIH 3T3 Cells , Neoplasms/metabolism , Protein Binding , Smoothened Receptor/chemistry , Smoothened Receptor/metabolism , Sulfonamides/metabolism
14.
Chem Biol Drug Des ; 97(3): 546-552, 2021 03.
Article in English | MEDLINE | ID: mdl-32946174

ABSTRACT

The hedgehog (Hh) signaling pathway is involved in diverse aspects of cellular events. Aberrant activation of Hh signaling pathway drives oncogenic transformation for a wide range of cancers, and it is therefore a promising target in cancer therapy. In the principle of association and ring-opening, we designed and synthesized a series of Hh signaling pathway inhibitors with phenyl imidazole scaffold, which were biologically evaluated in Gli-Luc reporter assay. Compound 25 was identified to possess high potency with nanomolar IC50 , and moreover, it preserved the inhibition against wild-type and drug-resistant Smo-overexpressing cells. A molecular modeling study of compound 25 expounded its binding mode to Smo receptor, providing a basis for the further structural modification of phenyl imidazole analogs.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Hedgehog Proteins/antagonists & inhibitors , Imidazoles/chemistry , Signal Transduction/drug effects , Anilides/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Hedgehog Proteins/metabolism , Humans , Imidazoles/metabolism , Imidazoles/pharmacology , Ligands , Molecular Docking Simulation , Pyridines/pharmacology , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Structure-Activity Relationship
15.
J Cell Biol ; 220(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-33258871

ABSTRACT

Primary cilia function as critical signaling hubs whose absence leads to severe disorders collectively known as ciliopathies; our knowledge of ciliogenesis remains limited. We show that Smo induces ciliogenesis through two distinct yet essential noncanonical Hh pathways in several cell types, including neurons. Surprisingly, ligand activation of Smo induces autophagy via an LKB1-AMPK axis to remove the satellite pool of OFD1. This is required, but not sufficient, for ciliogenesis. Additionally, Smo activates the Gαi-LGN-NuMA-dynein axis, causing accumulation of a portion of OFD1 at centrioles in early ciliogenesis. Both pathways are critical for redistribution of BBS4 from satellites to centrioles, which is also mediated by OFD1 centriolar translocation. Notably, different Smo agonists, which activate Smo distinctly, activate one or the other of these pathways; only in combination they recapitulate the activity of Hh ligand. These studies provide new insight into physiological stimuli (Hh) that activate autophagy and promote ciliogenesis and introduce a novel role for the Gαi-LGN-NuMA-dynein complex in this process.


Subject(s)
Autophagy , Cilia/metabolism , Hedgehog Proteins/metabolism , Organogenesis , Signal Transduction , AMP-Activated Protein Kinase Kinases , Adenylate Kinase/metabolism , Autophagy/drug effects , Basal Bodies/drug effects , Basal Bodies/metabolism , Cell Cycle Proteins/metabolism , Cells, Cultured , Centrioles/drug effects , Centrioles/metabolism , Cilia/drug effects , Dyneins/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Organogenesis/drug effects , Piperazines/pharmacology , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Proteins/metabolism , Proteolysis/drug effects , Pyridines/pharmacology , RNA, Small Interfering/metabolism , Retinal Pigment Epithelium/cytology , Serum/metabolism , Signal Transduction/drug effects , Smoothened Receptor/agonists , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/metabolism
17.
Expert Opin Ther Targets ; 24(11): 1159-1181, 2020 11.
Article in English | MEDLINE | ID: mdl-32990091

ABSTRACT

INTRODUCTION: Medulloblastoma (MB) is a heterogeneous tumor of the cerebellum that is divided into four main subgroups with distinct molecular and clinical features. Sonic Hedgehog MB (SHH-MB) is the most genetically understood and occurs predominantly in childhood. Current therapies consist of aggressive and non-targeted multimodal approaches that are often ineffective and cause long-term complications. These problems intensify the need to develop molecularly targeted therapies to improve outcome and reduce treatment-related morbidities. In this scenario, Hedgehog (HH) signaling, a developmental pathway whose deregulation is involved in the pathogenesis of several malignancies, has emerged as an attractive druggable pathway for SHH-MB therapy. AREAS COVERED: This review provides an overview of the advancements in the HH antagonist research field. We place an emphasis on Smoothened (SMO) and glioma-associated oncogene homolog (GLI) inhibitors and immunotherapy approaches that are validated in preclinical SHH-MB models and that have therapeutic potential for MB patients. Literature from Pubmed and data reported on ClinicalTrial.gov up to August 2020 were considered. EXPERT OPINION: Extensive-omics analysis has enhanced our knowledge and has transformed the way that MB is studied and managed. The clinical use of SMO antagonists has yet to be determined, however, future GLI inhibitors and multitargeting approaches are promising.


Subject(s)
Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , Molecular Targeted Therapy , Animals , Antineoplastic Agents/pharmacology , Cerebellar Neoplasms/pathology , Hedgehog Proteins/metabolism , Humans , Immunotherapy , Medulloblastoma/pathology , Signal Transduction/drug effects , Smoothened Receptor/antagonists & inhibitors , Zinc Finger Protein GLI1/antagonists & inhibitors
18.
Int J Mol Sci ; 21(18)2020 Sep 18.
Article in English | MEDLINE | ID: mdl-32962123

ABSTRACT

Smoothened (SMO) belongs to the Hedgehog (HH) signaling pathway, which regulates cell growth, migration, invasion and stem cells in cancer. The HH signaling pathway includes both canonical and noncanonical pathways. The canonical HH pathway functions through major HH molecules such as HH ligands, PTCH, SMO and GLI, whereas the noncanonical HH pathway involves the activation of SMO or GLI through other pathways. The role of SMO has been discussed in different types of cancer, including breast, liver, pancreatic and colon cancers. SMO expression correlates with tumor size, invasiveness, metastasis and recurrence. In addition, SMO inhibitors can suppress cancer formation, reduce the proliferation of cancer cells, trigger apoptosis and suppress cancer stem cell activity. A better understanding of the role of SMO in cancer could contribute to the development of novel therapeutic approaches.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Colonic Neoplasms/metabolism , Liver Neoplasms/metabolism , Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms/metabolism , Smoothened Receptor/antagonists & inhibitors , Animals , Breast Neoplasms/drug therapy , Colonic Neoplasms/drug therapy , Female , Hedgehog Proteins/metabolism , Humans , Liver Neoplasms/drug therapy , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/drug therapy , Signal Transduction/drug effects , Signal Transduction/genetics , Smoothened Receptor/metabolism
19.
Oncogene ; 39(42): 6589-6605, 2020 10.
Article in English | MEDLINE | ID: mdl-32929154

ABSTRACT

Triple-negative breast cancer (TNBC) and HER2-positive breast cancer are particularly aggressive and associated with unfavorable prognosis. TNBC lacks effective treatments. HER2-positive tumors have treatment options but often acquire resistance to HER2-targeted therapy after initial response. To address these challenges, we determined whether novel combinations of JAK2-STAT3 and SMO-GLI1/tGLI1 inhibitors synergistically target TNBC and HER2 breast cancer since these two pathways are concurrently activated in both tumor types and enriched in metastatic tumors. Herein, we show that novel combinations of JAK2 inhibitors (ruxolitinib and pacritinib) with SMO inhibitors (vismodegib and sonidegib) synergistically inhibited in vitro growth of TNBC and HER2-positive trastuzumab-resistant BT474-TtzmR cells. Synergy was also observed against breast cancer stem cells. To determine if the combination is efficacious in inhibiting metastasis, we treated mice with intracardially inoculated TNBC cells and found the combination to inhibit lung and liver metastases, and prolong host survival without toxicity. The combination inhibited orthotopic growth, VEGF-A expression, and tumor vasculature of both TNBC and HER2-positive trastuzumab-refractory breast cancer. Lung metastasis of orthotopic BT474-TtzmR xenografts was suppressed by the combination. Together, our results indicated that dual targeting of JAK2 and SMO resulted in synergistic suppression of breast cancer growth and metastasis, thereby supporting future clinical testing.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Janus Kinase 2/antagonists & inhibitors , Signal Transduction/drug effects , Smoothened Receptor/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Alternative Splicing , Anilides/pharmacology , Anilides/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Bridged-Ring Compounds/pharmacology , Bridged-Ring Compounds/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , Female , Humans , Janus Kinase 2/metabolism , Mice , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Nitriles , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Receptor, ErbB-2/metabolism , STAT3 Transcription Factor/metabolism , Smoothened Receptor/metabolism , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
20.
Bioorg Med Chem ; 28(16): 115584, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32690258

ABSTRACT

Triple-negative breast cancer (TNBC), a subset of breast cancers, have poorer survival than other breast cancer types. Recent studies have demonstrated that the abnormal Hedgehog (Hh) pathway is activated in TNBC and that these treatment-resistant cancers are sensitive to inhibition of the Hh pathway. Smoothened (Smo) protein is a vital constituent in Hh signaling and an attractive drug target. Vismodegib (VIS) is one of the most widely studied Smo inhibitors. But the clinical application of Smo inhibitors is limited to adult patients with BCC and AML, with many side effects. Therefore, it's necessary to develop novel Smo inhibitor with better profiles. Twenty [1,2,4]triazolo[4,3-a]pyridines were designed, synthesized and screened as Smo inhibitors. Four of these novel compounds showed directly bound to Smo protein with stronger binding affinity than VIS. The new compounds showed broad anti-proliferative activity against cancer cell lines in vitro, especially triple-negative breast cancer cells. Mechanistic studies demonstrated that TPB15 markedly induced cell cycle arrest and apoptosis in MDA-MB-468 cells. TPB15 blocked Smo translocation into the cilia and reduced Smo protein and mRNA expression. Furthermore, the expression of the downstream regulatory factor glioma-associated oncogene 1 (Gli1) was significantly inhibited. Finally, TPB15 demonstrated greater anti-tumor activity in our animal models than VIS with lower toxicity. Hence, these results support further optimization of this novel scaffold to develop improved Smo antagonists.


Subject(s)
Antineoplastic Agents/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Pyridines/pharmacology , Signal Transduction/drug effects , Smoothened Receptor/antagonists & inhibitors , Triazoles/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Female , Hedgehog Proteins/metabolism , Humans , Mice, Inbred BALB C , Pyridines/chemistry , Pyridines/therapeutic use , Smoothened Receptor/metabolism , Triazoles/chemistry , Triazoles/therapeutic use , Triple Negative Breast Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...