Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Bioeng Transl Med ; 8(5): e10541, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37693068

ABSTRACT

Multiple studies have shown that the progression of breast cancer depends on multiple signaling pathways, suggesting that therapies with multitargeted anticancer agents will offer improved therapeutic benefits through synergistic effects in inhibiting cancer growth. Dual-targeted inhibitors of phosphoinositide 3-kinase (PI3-K) and histone deacetylase (HDAC) have emerged as promising cancer therapy candidates. However, poor aqueous solubility and bioavailability limited their efficacy in cancer. The present study investigates the encapsulation of a PI3-Kδ/HDAC6 dual inhibitor into hybrid block copolymers (polylactic acid-methoxy polyethylene glycol; polylactic acid-polyethylene glycol-polypropylene glycol-polyethylene glycol-polylactic acid) (HSB-510) as a delivery system to target PI3-Kδ and HDAC6 pathways in breast cancer cells. The prepared HSB-510 showed an average diameter of 96 ± 3 nm, a zeta potential of -17 ± 2 mV, and PDI of ˂0.1 with a slow and sustained release profile of PI3-Kδ/HDAC6 inhibitors in a nonphysiological buffer. In vitro studies with HSB-510 have demonstrated substantial growth inhibition of breast cancer cell lines, MDA-MB-468, SUM-149, MCF-7, and Ehrlich ascites carcinoma (EAC) as well as downregulation of phospho-AKT, phospho-ERK, and c-Myc levels. Importantly, bi-weekly treatment of Balb/c wild-type mice harboring EAC cells with HSB-510 at a dose of 25 mg/kg resulted in significant tumor growth inhibition. The treatment with HSB-510 was without any significant effect on the body weights of the mice. These results demonstrate that a novel Quatramer encapsulation of a PI3-Kδ/HDAC6 dual inhibitor (HSB-510) represents an approach for the successful targeting of breast cancer and potentially other cancer types.

2.
PLoS Comput Biol ; 17(9): e1009450, 2021 09.
Article in English | MEDLINE | ID: mdl-34570764

ABSTRACT

Understanding relationships between spontaneous cancer in companion (pet) canines and humans can facilitate biomarker and drug development in both species. Towards this end we developed an experimental-bioinformatic protocol that analyzes canine transcriptomics data in the context of existing human data to evaluate comparative relevance of canine to human cancer. We used this protocol to characterize five canine cancers: melanoma, osteosarcoma, pulmonary carcinoma, B- and T-cell lymphoma, in 60 dogs. We applied an unsupervised, iterative clustering method that yielded five co-expression modules and found that each cancer exhibited a unique module expression profile. We constructed cancer models based on the co-expression modules and used the models to successfully classify the canine data. These canine-derived models also successfully classified human tumors representing the same cancers, indicating shared cancer biology between canines and humans. Annotation of the module genes identified cancer specific pathways relevant to cells-of-origin and tumor biology. For example, annotations associated with melanin production (PMEL, GPNMB, and BACE2), synthesis of bone material (COL5A2, COL6A3, and COL12A1), synthesis of pulmonary surfactant (CTSH, LPCAT1, and NAPSA), ribosomal proteins (RPL8, RPS7, and RPLP0), and epigenetic regulation (EDEM1, PTK2B, and JAK1) were unique to melanoma, osteosarcoma, pulmonary carcinoma, B- and T-cell lymphoma, respectively. In total, 152 biomarker candidates were selected from highly expressing modules for each cancer type. Many of these biomarker candidates are under-explored as drug discovery targets and warrant further study. The demonstrated transferability of classification models from canines to humans enforces the idea that tumor biology, biomarker targets, and associated therapeutics, discovered in canines, may translate to human medicine.


Subject(s)
Dog Diseases/genetics , Gene Regulatory Networks , Neoplasms/genetics , Neoplasms/veterinary , Animals , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/veterinary , Computational Biology , Dog Diseases/classification , Dogs , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/veterinary , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/veterinary , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/veterinary , Melanoma/genetics , Melanoma/veterinary , Molecular Sequence Annotation , Molecular Targeted Therapy , Neoplasms/classification , Oncogenes , Osteosarcoma/genetics , Osteosarcoma/veterinary , Species Specificity , Translational Research, Biomedical
3.
J Med Chem ; 63(8): 4256-4292, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32212730

ABSTRACT

A series of quinazolin-4-one based hydroxamic acids was rationally designed and synthesized as novel dual PI3K/HDAC inhibitors by incorporating an HDAC pharmacophore into a PI3K inhibitor (Idelalisib) via an optimized linker. Several of these dual inhibitors were highly potent (IC50 < 10 nM) and selective against PI3Kγ, δ and HDAC6 enzymes and exhibited good antiproliferative activity against multiple cancer cell lines. The lead compound 48c, induced necrosis in several mutant and FLT3-resistant AML cell lines and primary blasts from AML patients, while showing no cytotoxicity against normal PBMCs, NIH3T3, and HEK293 cells. Target engagement of PI3Kδ and HDAC6 by 48c was demonstrated in MV411 cells using the cellular thermal shift assay (CETSA). Compound 48c showed good pharmacokinetics properties in mice via intraperitoneal (ip) administration and provides a means to examine the biological effects of inhibiting these two important enzymes with a single molecule, either in vitro or in vivo.


Subject(s)
Drug Design , Histone Deacetylase Inhibitors/chemical synthesis , Hydroxamic Acids/chemical synthesis , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/chemical synthesis , Quinazolinones/chemical synthesis , Animals , Cell Line, Tumor , Drug Evaluation, Preclinical/methods , Female , HEK293 Cells , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Mice , Mice, Inbred BALB C , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Quinazolinones/pharmacology , Rats
4.
Drug Discov Today ; 24(12): 2341-2349, 2019 12.
Article in English | MEDLINE | ID: mdl-31585169

ABSTRACT

The National Center for Advancing Translational Sciences (NCATS) Pharmaceutical Collection (NPC), a comprehensive collection of clinically approved drugs, was made a public resource in 2011. Over the past decade, the NPC has been systematically profiled for activity across an array of pathways and disease models, generating an unparalleled amount of data. These data have not only enabled the identification of new repurposing candidates with several in clinical trials, but also uncovered new biological insights into drug targets and disease mechanisms. This retrospective provides an update on the NPC in terms of both successes and lessons learned. We also report our efforts in bringing the NPC up-to-date with drugs approved in recent years.


Subject(s)
Drug Approval , Drug Repositioning , National Center for Advancing Translational Sciences (U.S.)/trends , Drug Development/trends , Humans , United States
5.
J Med Chem ; 58(17): 7057-75, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26291341

ABSTRACT

We report here a novel series of benzimidazole sulfonamides that act as antagonists of the S1P1 receptor, identified by exploiting an understanding of the pharmacophore of a high throughput screening (HTS)-derived series of compounds described previously. Lead compound 2 potently inhibits S1P-induced receptor internalization in a cell-based assay (EC50 = 0.05 µM), but has poor physical properties and metabolic stability. Evolution of this compound through structure-activity relationship development and property optimization led to in vivo probes such as 4. However, this compound was unexpectedly found to be a potent CYP3A inducer in human hepatocytes, and thus further chemistry efforts were directed at addressing this liability. By employing a pregnane X receptor (PXR) reporter gene assay to prioritize compounds for further testing in human hepatocytes, we identified lipophilicity as a key molecular property influencing the likelihood of P450 induction. Ultimately, we have identified compounds such as 46 and 47, which demonstrate the desired S1P1 antagonist activity while having greatly reduced risk of CYP3A induction in humans. These compounds have excellent oral bioavailability in preclinical species and exhibit pharmacodynamic effects of S1P1 antagonism in several in vivo models following oral dosing. Relatively modest antitumor activity was observed in multiple xenograft models, however, suggesting that selective S1P1 antagonists would have limited utility as anticancer therapeutics as single agents.


Subject(s)
Benzimidazoles/chemistry , Pyridines/chemistry , Receptors, Lysosphingolipid/antagonists & inhibitors , Sulfonamides/chemistry , Administration, Oral , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Biological Availability , Cells, Cultured , Cytochrome P-450 CYP3A/biosynthesis , Cytochrome P-450 CYP3A Inducers/chemical synthesis , Cytochrome P-450 CYP3A Inducers/chemistry , Cytochrome P-450 CYP3A Inducers/pharmacology , Genes, Reporter , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Models, Molecular , Molecular Structure , Pregnane X Receptor , Pyridines/chemical synthesis , Pyridines/pharmacology , Receptors, Steroid/genetics , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays
6.
Bioorg Med Chem Lett ; 25(10): 2041-5, 2015.
Article in English | MEDLINE | ID: mdl-25890801

ABSTRACT

We have discovered a novel class of heterocyclic sulfonamides that act as antagonists of the S1P1 receptor. While members of this series identified from a high-throughput screen showed promising levels of potency in a cell-based assay measuring the inhibition of receptor internalization, most compounds were excessively lipophilic and contained an oxidation-prone thioether moiety. As a result, such compounds suffered from poor physical properties and metabolic stability, limiting their utility as in vivo probes. By removing the thioether group and systematically developing an understanding of structure-activity relationships and the effects of lipophilicity on potency within this series, we have been able to identify potent compounds with vastly improved physical properties. A representative enantiopure triazole sulfonamide (33) has measurable bioavailability following a low (3mg/kg) oral dose in rat, highlighting an achievement of the early hit-to-lead efforts for this series.


Subject(s)
Drug Discovery , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/pharmacology , Receptors, Lysosphingolipid/antagonists & inhibitors , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology , Animals , Heterocyclic Compounds/chemistry , Protein Binding/drug effects , Rats , Structure-Activity Relationship , Sulfonamides/chemistry
7.
J Med Chem ; 57(23): 9958-70, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25458601

ABSTRACT

KIFC1 (HSET), a member of the kinesin-14 family of motor proteins, plays an essential role in centrosomal bundling in cancer cells, but its function is not required for normal diploid cell division. To explore the potential of KIFC1 as a therapeutic target for human cancers, a series of potent KIFC1 inhibitors featuring a phenylalanine scaffold was developed from hits identified through high-throughput screening (HTS). Optimization of the initial hits combined both design-synthesis-test cycles and an integrated high-throughput synthesis and biochemical screening method. An important aspect of this integrated method was the utilization of DMSO stock solutions of compounds registered in the corporate compound collection as synthetic reactants. Using this method, over 1500 compounds selected for structural diversity were quickly assembled in assay-ready 384-well plates and were directly tested after the necessary dilutions. Our efforts led to the discovery of a potent KIFC1 inhibitor, AZ82, which demonstrated the desired centrosome declustering mode of action in cell studies.


Subject(s)
Alanine/analogs & derivatives , Kinesins/antagonists & inhibitors , Pyridines/chemical synthesis , Alanine/chemical synthesis , Alanine/pharmacology , Animals , HeLa Cells , High-Throughput Screening Assays/methods , Humans , Inhibitory Concentration 50 , Mice , Phenylalanine/analogs & derivatives , Pyridines/pharmacology , Rats , Structure-Activity Relationship
8.
Pharmaceut Med ; 28(1): 1-6, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24855373

ABSTRACT

While there are approximately 7,000 identified human diseases considered as "rare" based on population prevalence or incidence, the cumulative impact runs into the millions of patients globally. Although the genetic underpinnings of more than 2,000 rare diseases have been elucidated, there remains a paucity of therapeutic options, frequently due to lack of commercial interest. Development programs suffer high attrition within the so-called "Valley of Death," in which the risks of scientific failure are still too high to justify the increasing development costs. This problem is common to any drug development campaign, but it is particularly exacerbated in the rare diseases, many of which arise in childhood. To stimulate development of therapeutics for these otherwise underserved patient populations, a number of regulatory incentives and research initiatives have been established. Extended patent protections, expedited regulatory reviews for qualified drug sponsors, and clinical trial grant support aim to foster interest in completing development programs. To stimulate researchers to embark on rare disease drug development campaigns, earlier-stage preclinical research resources have been created, as well, such as the Therapeutics for Rare and Neglected Diseases (TRND) program at the U.S. National Institutes of Health (NIH). TRND is a unique NIH program created to support drug development through formation of public-private partnerships. These partnerships leverage the robust biopharmaceutical industry experience of the TRND staff scientists and the deep disease area expertise of the collaborating partners. Each project adopted into the TRND portfolio aims to satisfy two broad goals: developing a novel therapy for a rare or otherwise neglected disease, and exploring ways to accelerate the drug development process overall so that lessons learned can be disseminated to the wider community undertaking translational research. This article discusses common obstacles and opportunities for therapeutic development, and provides examples of the types of projects TRND has undertaken across a broad range of pediatric rare disorders.

9.
Bioorg Med Chem Lett ; 14(9): 2265-8, 2004 May 03.
Article in English | MEDLINE | ID: mdl-15081022

ABSTRACT

A series of novel compounds with both 5-lipoxygenase (5-LO) inhibitory and histamine H(1) receptor antagonist activity were designed for the treatment of asthma. These dual-function compounds were made by connecting 5-LO and H(1) pharmacophores,N-hydroxyureas and benzhydryl piperazines, respectively. A range of in vitro activities was observed, with the furan analog 10 demonstrating both activities in an animal model. The activities observed were compared to single-function drugs.


Subject(s)
Histamine H1 Antagonists/pharmacology , Lipoxygenase Inhibitors , Lipoxygenase Inhibitors/pharmacology , Animals , Caco-2 Cells , Histamine H1 Antagonists/chemistry , Histamine H1 Antagonists/pharmacokinetics , Humans , Lipoxygenase Inhibitors/chemistry , Lipoxygenase Inhibitors/pharmacokinetics , Microsomes, Liver/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL