1
|
Bertulfo K, Perez-Duran P, Miller H, Ma C, Ambesi-Impiombato A, Samon J, Mackey A, Lin WHW, Ferrando AA, Palomero T. Therapeutic targeting of the NOTCH1 and neddylation pathways in T cell acute lymphoblastic leukemia. Proc Natl Acad Sci U S A 2025; 122:e2426742122. [PMID: 40163723 PMCID: PMC12002235 DOI: 10.1073/pnas.2426742122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 01/30/2025] [Indexed: 04/02/2025] Open
Abstract
Gamma Secretase Inhibitors (GSIs) effectively block oncogenic Notch homolog-1 (NOTCH1), a characteristic feature of T cell acute lymphoblastic leukemias (T-ALL). However, their clinical application has been stalled by the induction of severe gastrointestinal toxicity resulting from the inhibition of NOTCH signaling in the gut, which translates into increased goblet cell differentiation. Genome-wide CRISPR loss-of-function screen in the colon cancer cell line LS174T identified the neddylation pathway as a main regulator of goblet cell differentiation upon NOTCH1 inhibition. Consistently, pharmacologic inhibition of the neddylation pathway with the small molecule inhibitor MLN4924, rescued GSI-induced differentiation in LS174T cells. Mechanistically, neddylation inhibition by MLN4924 increases the protein stability of Hairy and enhancer of split-1, a direct NOTCH1 transcriptional target and key regulator of absorptive and secretory cell fate decisions. Combined treatment with GSI and MLN4924 in a murine Notch1-dependent model of T-ALL led to leukemia regression and improved overall survival in the absence of gut toxicity. Overall, these results support the combined targeting of the NOTCH1 and neddylation pathways for the treatment of NOTCH1-induced T-ALL.
Collapse
Affiliation(s)
- Kalay Bertulfo
- Institute for Cancer Genetics, Columbia University, New York, NY10032
- Department of Biological Sciences, Columbia University, New York, NY10027
| | - Pablo Perez-Duran
- Institute for Cancer Genetics, Columbia University, New York, NY10032
| | - Hannah Miller
- Institute for Cancer Genetics, Columbia University, New York, NY10032
| | - Cindy Ma
- Institute for Cancer Genetics, Columbia University, New York, NY10032
| | | | - Jeremy Samon
- Institute for Cancer Genetics, Columbia University, New York, NY10032
| | - Adam Mackey
- Institute for Cancer Genetics, Columbia University, New York, NY10032
| | - Wen-Hsuan Wendy Lin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY10032
| | - Adolfo A. Ferrando
- Institute for Cancer Genetics, Columbia University, New York, NY10032
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY10032
- Department of Pediatrics, Columbia University Medical Center, New York, NY10032
| | - Teresa Palomero
- Institute for Cancer Genetics, Columbia University, New York, NY10032
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY10032
| |
Collapse
|
2
|
Jerom JP, Jalal A, Sajan AL, Soman R, Nair RH, Narayanan SP. In-vitro Neuro-2a cytotoxicity analysis and molecular docking investigation on potential anti-amyloid agents from Adiantum lunulatum. Heliyon 2024; 10:e38127. [PMID: 39381205 PMCID: PMC11458992 DOI: 10.1016/j.heliyon.2024.e38127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/22/2024] [Accepted: 09/18/2024] [Indexed: 10/10/2024] Open
Abstract
In neurodegenerative diseases, amyloid formation by some proteins cause neuronal damage and loss. To prevent this neuronal damage and loss certain pharmaceuticals are available. Many of these pharmaceuticals act on the neurodegenerative disease symptoms but not on the root cause. This study helps to detect more effective agents which directly act on the root cause and reduce the risk of neurodegenerative diseases. To identify new anti-amyloid agents, the folk medicinally important plant Adiantum lunulatum was collected, authenticated, dried, extracted with ethanol and analyzed by GC-MS method. The screening of the identified phytochemicals was done using the webservers swissADME and ProTox-II. In-vitro MTT assay using Neuro-2a cell lines was carried out to determine the cytotoxicity of the extract. The interactions of these phytochemicals with the amyloid forming peptides and proteins were predicted using the molecular docking tools such as AutoDock Vina and BIOVIA discovery studio visualizer 2020. Through GC-MS analysis, 18 different volatile phytochemicals were identified from the ethanol extract. From this, 7 phytochemicals were selected based on the computational non-toxicity prediction. In-vitro cytotoxicity analysis of the ethanol extract using Neuro-2a cell lines detected the IC50 value of 0.09 mg/ml. Of these, the phytochemical P1 (trans, trans-9, 12-Octadecadienoic acid, propyl ester) interacts with tau, and huntingtin proteins, P2 (2-Pentadecanone, 6, 10, 14-trimethyl-) interacts with prion protein. The phytochemicals P1, P3 (Ethyl oleate), P4 (Octadecanoic acid, ethyl ester), and P5 (Phytol) interact with acetylcholinesterase. P2, P4, P5 and P6 (Henicosanal), interact with BACE-1. The phytochemical P3 interacts with γ- Secretase. The interaction of P2 and P5 with BACE-1 and P3 with γ- Secretase show better inhibition in inhibitory constant (K i ) analysis. These phytochemicals have been predicted to show significant potential against the formation or breakdown of peptide/protein amyloids, and further in-vitro studies are necessary to develop them into anti-amyloid agents.
Collapse
Affiliation(s)
| | - Ajmal Jalal
- School of Biosciences, Mahatma Gandhi University, Kottayam, 686560, Kerala, India
| | - Ann Liya Sajan
- School of Biosciences, Mahatma Gandhi University, Kottayam, 686560, Kerala, India
| | - Reshma Soman
- School of Biosciences, Mahatma Gandhi University, Kottayam, 686560, Kerala, India
| | | | - Sunilkumar Puthenpurackal Narayanan
- NMR Facility, Institute for Integrated Programmes and Research in Basic Sciences. Mahatma Gandhi University, Kottayam, 686560, Kerala, India
| |
Collapse
|
3
|
Afjadi MN, Dabirmanesh B, Uversky VN. Therapeutic approaches in proteinopathies. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 206:341-388. [PMID: 38811085 DOI: 10.1016/bs.pmbts.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
A family of maladies known as amyloid disorders, proteinopathy, or amyloidosis, are characterized by the accumulation of abnormal protein aggregates containing cross-β-sheet amyloid fibrils in many organs and tissues. Often, proteins that have been improperly formed or folded make up these fibrils. Nowadays, most treatments for amyloid illness focus on managing symptoms rather than curing or preventing the underlying disease process. However, recent advances in our understanding of the biology of amyloid diseases have led to the development of innovative therapies that target the emergence and accumulation of amyloid fibrils. Examples of these treatments include the use of small compounds, monoclonal antibodies, gene therapy, and others. In the end, even if the majority of therapies for amyloid diseases are symptomatic, greater research into the biology behind these disorders is identifying new targets for potential therapy and paving the way for the development of more effective treatments in the future.
Collapse
Affiliation(s)
- Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Vladimir N Uversky
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute for Biological Instrumentation, Pushchino, Moscow, Russia; Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.
| |
Collapse
|
4
|
Ibrahim R, Assi T, Khoury R, Ngo C, Faron M, Verret B, Lévy A, Honoré C, Hénon C, Le Péchoux C, Bahleda R, Le Cesne A. Desmoid-type fibromatosis: Current therapeutic strategies and future perspectives. Cancer Treat Rev 2024; 123:102675. [PMID: 38159438 DOI: 10.1016/j.ctrv.2023.102675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Desmoid tumors (DT) are rare, slow-growing, locally invasive soft tissue tumors that often pose significant therapeutic challenges. Traditional management strategies including active surveillance, surgery, radiotherapy, and systemic therapy which are associated with varying recurrence rates and high morbidity. Given the challenging nature of DT and the modest outcomes associated with current treatment strategies, there has been a growing interest in the field of γ-secretase inhibitors as a result of its action on the Wnt/β-catenin signaling pathway. In this review article, we will shed the light on the pathogenesis and molecular biology of DT, discuss its symptoms and diagnosis, and provide a comprehensive review of the traditional therapeutic approaches. We will also delve into the mechanisms of action of γ-secretase inhibitors, its efficacy, and the existing preclinical and clinical data available to date on the use of these agents, as well as the potential challenges and future prospects in the treatment landscape of these tumors.
Collapse
Affiliation(s)
- Rebecca Ibrahim
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France
| | - Tarek Assi
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France; Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France.
| | - Rita Khoury
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France
| | - Carine Ngo
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Matthieu Faron
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Benjamin Verret
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Antonin Lévy
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Charles Honoré
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Clémence Hénon
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | | | | | - Axel Le Cesne
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France; Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
5
|
Nystuen KL, McNamee SM, Akula M, Holton KM, DeAngelis MM, Haider NB. Alzheimer's Disease: Models and Molecular Mechanisms Informing Disease and Treatments. Bioengineering (Basel) 2024; 11:45. [PMID: 38247923 PMCID: PMC10813760 DOI: 10.3390/bioengineering11010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Alzheimer's Disease (AD) is a complex neurodegenerative disease resulting in progressive loss of memory, language and motor abilities caused by cortical and hippocampal degeneration. This review captures the landscape of understanding of AD pathology, diagnostics, and current therapies. Two major mechanisms direct AD pathology: (1) accumulation of amyloid β (Aβ) plaque and (2) tau-derived neurofibrillary tangles (NFT). The most common variants in the Aβ pathway in APP, PSEN1, and PSEN2 are largely responsible for early-onset AD (EOAD), while MAPT, APOE, TREM2 and ABCA7 have a modifying effect on late-onset AD (LOAD). More recent studies implicate chaperone proteins and Aβ degrading proteins in AD. Several tests, such as cognitive function, brain imaging, and cerebral spinal fluid (CSF) and blood tests, are used for AD diagnosis. Additionally, several biomarkers seem to have a unique AD specific combination of expression and could potentially be used in improved, less invasive diagnostics. In addition to genetic perturbations, environmental influences, such as altered gut microbiome signatures, affect AD. Effective AD treatments have been challenging to develop. Currently, there are several FDA approved drugs (cholinesterase inhibitors, Aß-targeting antibodies and an NMDA antagonist) that could mitigate AD rate of decline and symptoms of distress.
Collapse
Affiliation(s)
- Kaden L. Nystuen
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Shannon M. McNamee
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Monica Akula
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Kristina M. Holton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Margaret M. DeAngelis
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Neena B. Haider
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Ashour MM, Mabrouk M, Aboelnasr MA, Beherei HH, Tohamy KM, Das DB. Anti-Obesity Drug Delivery Systems: Recent Progress and Challenges. Pharmaceutics 2023; 15:2635. [PMID: 38004612 PMCID: PMC10674714 DOI: 10.3390/pharmaceutics15112635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Obesity has reached an epidemic proportion in the last thirty years, and it is recognized as a major health issue in modern society now with the possibility of serious social and economic consequences. By the year 2030, nearly 60% of the global population may be obese or overweight, which emphasizes a need for novel obesity treatments. Various traditional approaches, such as pharmacotherapy and bariatric surgery, have been utilized in clinical settings to treat obesity. However, these methods frequently show the possibility of side effects while remaining ineffective. There is, therefore, an urgent need for alternative obesity treatments with improved efficacy and specificity. Polymeric materials and chemical strategies are employed in emerging drug delivery systems (DDSs) to enhance therapy effectiveness and specificity by stabilizing and controlling the release of active molecules such as natural ingredients. Designing DDSs is currently a top priority research objective with an eye towards creating obesity treatment approaches. In reality, the most recent trends in the literature demonstrate that there are not enough in-depth reviews that emphasize the current knowledge based on the creation and design of DDSs for obesity treatment. It is also observed in the existing literature that a complex interplay of different physical and chemical parameters must be considered carefully to determine the effectiveness of the DDSs, including microneedles, for obesity treatment. Additionally, it is observed that these properties depend on how the DDS is synthesized. Although many studies are at the animal-study stage, the use of more advanced DDS techniques would significantly enhance the development of safe and efficient treatment approaches for obese people in the future. Considering these, this review provides an overview of the current anti-obesity treatment approaches as well as the conventional anti-obesity therapeutics. The article aims to conduct an in-depth discussion on the current trends in obesity treatment approaches. Filling in this knowledge gap will lead to a greater understanding of the safest ways to manage obesity.
Collapse
Affiliation(s)
- Mohamed M. Ashour
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo 11829, Egypt;
| | - Mostafa Mabrouk
- Refractories, Ceramics and Building Materials Department, National Research Centre, 33 El Bohouth St., Dokki, Giza 12622, Egypt;
| | - Mohamed A. Aboelnasr
- Biophysics Branch, Faculty of Science, Al-Azhar University, Nasr City, Cairo 11884, Egypt; (M.A.A.); (K.M.T.)
| | - Hanan H. Beherei
- Refractories, Ceramics and Building Materials Department, National Research Centre, 33 El Bohouth St., Dokki, Giza 12622, Egypt;
| | - Khairy M. Tohamy
- Biophysics Branch, Faculty of Science, Al-Azhar University, Nasr City, Cairo 11884, Egypt; (M.A.A.); (K.M.T.)
| | - Diganta B. Das
- Department of Chemical Engineering, Loughborough University, Loughborough LE113TU, UK
| |
Collapse
|
7
|
McCaw TR, Inga E, Chen H, Jaskula‐Sztul R, Dudeja V, Bibb JA, Ren B, Rose JB. Gamma Secretase Inhibitors in Cancer: A Current Perspective on Clinical Performance. Oncologist 2021; 26:e608-e621. [PMID: 33284507 PMCID: PMC8018325 DOI: 10.1002/onco.13627] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/13/2020] [Indexed: 01/01/2023] Open
Abstract
Gamma secretase inhibitors (GSIs), initially developed as Alzheimer's therapies, have been repurposed as anticancer agents given their inhibition of Notch receptor cleavage. The success of GSIs in preclinical models has been ascribed to induction of cancer stem-like cell differentiation and apoptosis, while also impairing epithelial-to-mesenchymal transition and sensitizing cells to traditional chemoradiotherapies. The promise of these agents has yet to be realized in the clinic, however, as GSIs have failed to demonstrate clinical benefit in most solid tumors with the notable exceptions of CNS malignancies and desmoid tumors. Disappointing clinical performance to date reflects important questions that remain to be answered. For example, what is the net impact of these agents on antitumor immune responses, and will they require concurrent targeting of tumor-intrinsic compensatory pathways? Addressing these limitations in our current understanding of GSI mechanisms will undoubtedly facilitate their rational incorporation into combinatorial strategies and provide a valuable tool with which to combat Notch-dependent cancers. In the present review, we provide a current understanding of GSI mechanisms, discuss clinical performance to date, and suggest areas for future investigation that might maximize the utility of these agents. IMPLICATIONS FOR PRACTICE: The performance of gamma secretase inhibitors (GSIs) in clinical trials generally has not reflected their encouraging performance in preclinical studies. This review provides a current perspective on the clinical performance of GSIs across various solid tumor types alongside putative mechanisms of antitumor activity. Through exploration of outstanding gaps in knowledge as well as reasons for success in certain cancer types, the authors identify areas for future investigation that will likely enable incorporation of GSIs into rational combinatorial strategies for superior tumor control and patient outcomes.
Collapse
Affiliation(s)
- Tyler R. McCaw
- Divisions of Surgical Oncology, The University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Evelyn Inga
- Divisions of Surgical Oncology, The University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Herbert Chen
- Breast & Endocrine Surgery, The University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Renata Jaskula‐Sztul
- Breast & Endocrine Surgery, The University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Vikas Dudeja
- Divisions of Surgical Oncology, The University of Alabama at BirminghamBirminghamAlabamaUSA
| | - James A. Bibb
- Gastrointestinal Surgery, The University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Bin Ren
- Vascular Surgery & Endovascular Therapy, Department of Surgery, The University of Alabama at BirminghamBirminghamAlabamaUSA
| | - J. Bart Rose
- Divisions of Surgical Oncology, The University of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
8
|
Zhu H, Dronamraju V, Xie W, More SS. Sulfur-containing therapeutics in the treatment of Alzheimer's disease. Med Chem Res 2021; 30:305-352. [PMID: 33613018 PMCID: PMC7889054 DOI: 10.1007/s00044-020-02687-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/06/2020] [Indexed: 12/12/2022]
Abstract
Sulfur is widely existent in natural products and synthetic organic compounds as organosulfur, which are often associated with a multitude of biological activities. OBenzothiazole, in which benzene ring is fused to the 4,5-positions of the thiazolerganosulfur compounds continue to garner increasing amounts of attention in the field of medicinal chemistry, especially in the development of therapeutic agents for Alzheimer's disease (AD). AD is a fatal neurodegenerative disease and the primary cause of age-related dementia posing severe societal and economic burdens. Unfortunately, there is no cure for AD. A lot of research has been conducted on sulfur-containing compounds in the context of AD due to their innate antioxidant potential and some are currently being evaluated in clinical trials. In this review, we have described emerging trends in the field, particularly the concept of multi-targeting and formulation of disease-modifying strategies. SAR, pharmacological targets, in vitro/vivo ADMET, efficacy in AD animal models, and applications in clinical trials of such sulfur compounds have also been discussed. This article provides a comprehensive review of organosulfur-based AD therapeutic agents and provides insights into their future development.
Collapse
Affiliation(s)
- Haizhou Zhu
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Venkateshwara Dronamraju
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Wei Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Swati S. More
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
9
|
Tyagi A, Chandrasekaran B, Kolluru V, Baby BV, Sripathi CA, Ankem MK, Ramisetti SR, Chirasani VR, Dokholyan NV, Sharma AK, Damodaran C. ASR490, a Small Molecule, Overrides Aberrant Expression of Notch1 in Colorectal Cancer. Mol Cancer Ther 2020; 19:2422-2431. [PMID: 33087513 PMCID: PMC10694926 DOI: 10.1158/1535-7163.mct-19-0949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 05/14/2020] [Accepted: 10/05/2020] [Indexed: 11/16/2022]
Abstract
Notch1 activation triggers significant oncogenic signaling that manifests as enhanced metastatic potential and tumorigenesis in colorectal cancer. Novel small-molecule inhibitors, mainly plant-derived analogs, have low toxicity profiles and higher bioavailability. In this study, we have developed a small molecule, ASR490, by modifying structure of naturally occurring compound Withaferin A. ASR490 showed a growth-inhibitory potential by downregulating Notch1 signaling in HCT116 and SW620 cell lines. Docking studies and thermal shift assays confirmed that ASR490 binds to Notch1, whereas no changes in Notch2 and Notch3 expression were seen in colorectal cancer cells. Notch1 governs epithelial-to-mesenchymal transition signaling and is responsible for metastasis, which was abolished by ASR490 treatment. To further confirm the therapeutic potential of ASR490, we stably overexpressed Notch1 in HCT-116 cells and determined its inhibitory potential in transfected colorectal cancer (Notch1/HCT116) cells. ASR490 effectively prevented cell growth in both the vector (P = 0.005) and Notch1 (P = 0.05) transfectants. The downregulation of Notch1 signaling was evident, which corresponded with downregulation of mesenchymal markers, including N-cadherin and β-catenin and induction of E-cadherin in HCT-116 transfectants. Intraperitoneal administration of a 1% MTD dose of ASR490 (5 mg/kg) effectively suppressed the tumor growth in control (pCMV/HCT116) and Notch1/HCT116 in xenotransplanted mice. In addition, downregulation of Notch1 and survival signaling in ASR-treated tumors confirmed the in vitro results. In conclusion, ASR490 appears to be a potent agent that can inhibit Notch1 signaling in colorectal cancer.
Collapse
Affiliation(s)
- Ashish Tyagi
- Department of Urology, University of Louisville, Louisville, Kentucky
| | | | - Venkatesh Kolluru
- Department of Urology, University of Louisville, Louisville, Kentucky
| | - Becca V Baby
- Department of Urology, University of Louisville, Louisville, Kentucky
| | - Cibi A Sripathi
- Department of Urology, University of Louisville, Louisville, Kentucky
| | - Murali K Ankem
- Department of Urology, University of Louisville, Louisville, Kentucky
| | - Srinivasa R Ramisetti
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, Pennsylvania
| | - Venkat R Chirasani
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, Pennsylvania
| | - Nikolay V Dokholyan
- Department of Pharmacology, Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, Pennsylvania
| | - Chendil Damodaran
- Department of Urology, University of Louisville, Louisville, Kentucky.
| |
Collapse
|
10
|
Human-Induced Pluripotent Stem Cells and Herbal Small-Molecule Drugs for Treatment of Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21041327. [PMID: 32079110 PMCID: PMC7072986 DOI: 10.3390/ijms21041327] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by extracellular amyloid plaques composed of the β-amyloid peptides and intracellular neurofibrillary tangles and associates with progressive declines in memory and cognition. Several genes play important roles and regulate enzymes that produce a pathological accumulation of β-amyloid in the brain, such as gamma secretase (γ-secretase). Induced pluripotent stem cells from patients with Alzheimer’s disease with different underlying genetic mechanisms may help model different phenotypes of Alzheimer’s disease and facilitate personalized drug screening platforms for the identification of small molecules. We also discuss recent developments by γ-secretase inhibitors and modulators in the treatment of AD. In addition, small-molecule drugs isolated from Chinese herbal medicines have been shown effective in treating Alzheimer’s disease. We propose a mechanism of small-molecule drugs in treating Alzheimer’s disease. Combining therapy with different small-molecule drugs may increase the chance of symptomatic treatment. A customized strategy tailored to individuals and in combination with therapy may be a more suitable treatment option for Alzheimer’s disease in the future.
Collapse
|
11
|
Huang D, Deng M, Kuang S. Polymeric Carriers for Controlled Drug Delivery in Obesity Treatment. Trends Endocrinol Metab 2019; 30:974-989. [PMID: 31668904 PMCID: PMC6927547 DOI: 10.1016/j.tem.2019.09.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 02/08/2023]
Abstract
The global rise in the prevalence of obesity and affiliated metabolic syndrome poses a significant threat to human health. Various approaches, including bariatric surgery and pharmacotherapy, have been used in the clinical setting for obesity treatment; however, these conventional options remain ineffective and carry risks of adverse effects. Therefore, treatments with higher efficacy and specificity are urgently required. Emerging drug delivery systems use polymeric materials and chemical strategies to improve therapeutic efficacy and specificity through stabilization and spatiotemporally controlled release of antiobesity agents. In this review, we provide insights into current treatments for obesity with a focus on recent developments of polymeric carriers for enhanced antiobesity drug delivery.
Collapse
Affiliation(s)
- Di Huang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA; Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, USA; Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA
| | - Meng Deng
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, USA; Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA; School of Materials Engineering, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA; Center for Cancer Research, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
12
|
Ahn JE, Carrieri C, Dela Cruz F, Fullerton T, Hajos-Korcsok E, He P, Kantaridis C, Leurent C, Liu R, Mancuso J, Mendes da Costa L, Qiu R. Pharmacokinetic and Pharmacodynamic Effects of a γ-Secretase Modulator, PF-06648671, on CSF Amyloid-β Peptides in Randomized Phase I Studies. Clin Pharmacol Ther 2019; 107:211-220. [PMID: 31314925 PMCID: PMC6977340 DOI: 10.1002/cpt.1570] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/08/2019] [Indexed: 01/04/2023]
Abstract
γ‐Secretase modulators (GSMs) represent a promising therapy for Alzheimer's disease by reducing pathogenic amyloid‐β (Aβ) peptide production. Three phase I studies (NCT02316756, NCT02407353, and NCT02440100) investigated the safety/tolerability, pharmacokinetics (PKs), and pharmacodynamics (PDs) of the oral GSM, PF‐06648671. A PK/PD indirect‐response model was developed (using biomarker data) to simultaneously characterize differential effects of PF‐06648671 on multiple Aβ species in cerebrospinal fluid (CSF). Healthy subjects (n = 120) received single doses or multiple‐ascending doses of PF‐06648671/placebo for 14 days. No serious adverse events occurred; severe adverse eventswere deemed not drug related. PF‐06648671 decreased Aβ42 and Aβ40 concentrations in CSF, with greater effects on Aβ42, and increased Aβ37 and Aβ38 levels, particularly Aβ37. No significant change in total Aβ was observed. The PK/PD model well described the tendency of observed CSF Aβ data and the steady‐state effects of PF‐06648671, supporting its use for predicting central Aβ effects and optimal dose selection for GSMs in future trials.
Collapse
Affiliation(s)
| | | | | | | | - Eva Hajos-Korcsok
- Pfizer Inc, Cambridge, Massachusetts, USA.,Sunovion Pharmaceuticals, Marlborough, Massachusetts, USA
| | - Ping He
- Pfizer Inc, Cambridge, Massachusetts, USA.,Biogen Inc, Cambridge, Massachusetts, USA
| | | | - Claire Leurent
- Pfizer Inc, Cambridge, Massachusetts, USA.,Samsung Ventures America, Boston, Massachusetts, USA
| | - Richann Liu
- Pfizer Inc, Cambridge, Massachusetts, USA.,ICON, Boston, Massachusetts, USA
| | | | | | - Ruolun Qiu
- Pfizer Inc, Cambridge, Massachusetts, USA
| |
Collapse
|
13
|
Ryu JS, Robinson L, Raucher D. Elastin-Like Polypeptide Delivers a Notch Inhibitory Peptide to Inhibit Tumor Growth in Combination with Paclitaxel. J Chemother 2019; 31:23-29. [DOI: 10.1080/1120009x.2018.1537554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Jung Su Ryu
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Leslie Robinson
- Department of Chemistry and Physics, Belhaven University, Jackson, MississippiUSA
| | - Drazen Raucher
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
14
|
Li Y, Wu L, Yu M, Yang F, Wu B, Lu S, Tu M, Xu H. HIF-1α is Critical for the Activation of Notch Signaling in Neurogenesis During Acute Epilepsy. Neuroscience 2018; 394:206-219. [PMID: 30394322 DOI: 10.1016/j.neuroscience.2018.10.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 09/30/2018] [Accepted: 10/22/2018] [Indexed: 01/01/2023]
Abstract
Emerging evidence suggests that hypoxia-inducible factors (specifically, HIF-1α) and Notch signaling are involved in epileptogenesis and that cross-coupling exists between HIF-1α and Notch signaling in other diseases, including tumors and ischemia. However, the exact molecular mechanisms by which HIF-1α and Notch signaling affect the development of epilepsy, especially regarding neurogenesis, remain unclear. In the present study, we investigated the role of HIF-1α in neurogenesis and whether Notch signaling is involved in this process during epileptogenesis by assessing hippocampal apoptosis, neuronal injury, and the proliferation and differentiation of neural stem cells (NSCs) in four groups, including control, epilepsy, epilepsy+2-methoxyestradiol (2ME2) and epilepsy+GSI-IX (DAPT) groups. Our data demonstrated that HIF-1α mediated neurogenesis during acute epilepsy, which required the participation of Notch signaling. The immunoprecipitation data illustrated that HIF-1α activated Notch signaling by physically interacting with the Notch intracellular domain (NICD) in epilepsy. In conclusion, our results suggested that HIF-1α-Notch signaling enhanced neurogenesis in acute epilepsy and that neurogenesis during epileptogenesis was reduced once this pathway was blocked; thus, members of this pathway might be potential therapeutic targets for epilepsy.
Collapse
Affiliation(s)
- Yushuang Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, PR China
| | - Lei Wu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, PR China
| | - Minhua Yu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, PR China
| | - Fei Yang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, PR China
| | - Bo Wu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, PR China
| | - Shuting Lu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, PR China
| | - Mengqi Tu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, PR China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, PR China.
| |
Collapse
|
15
|
Perron A, Nishikawa Y, Iwata J, Shimojo H, Takaya J, Kobayashi K, Imayoshi I, Mbenza NM, Takenoya M, Kageyama R, Kodama Y, Uesugi M. Small-molecule screening yields a compound that inhibits the cancer-associated transcription factor Hes1 via the PHB2 chaperone. J Biol Chem 2018. [PMID: 29523683 DOI: 10.1074/jbc.ra118.002316] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The transcription factor Hes family basic helix-loop-helix transcription factor 1 (Hes1) is a downstream effector of Notch signaling and plays a crucial role in orchestrating developmental processes during the embryonic stage. However, its aberrant signaling in adulthood is linked to the pathogenesis of cancer. In the present study, we report the discovery of small organic molecules (JI051 and JI130) that impair the ability of Hes1 to repress transcription. Hes1 interacts with the transcriptional corepressor transducing-like enhancer of split 1 (TLE1) via an interaction domain comprising two tryptophan residues, prompting us to search a chemical library of 1,800 small molecules enriched for indole-like π-electron-rich pharmacophores for a compound that blocks Hes1-mediated transcriptional repression. This screening identified a lead compound whose extensive chemical modification to improve potency yielded JI051, which inhibited HEK293 cell proliferation with an EC50 of 0.3 μm Unexpectedly, using immunomagnetic isolation and nanoscale LC-MS/MS, we found that JI051 does not bind TLE1 but instead interacts with prohibitin 2 (PHB2), a cancer-associated protein chaperone. We also found that JI051 stabilizes PHB2's interaction with Hes1 outside the nucleus, inducing G2/M cell-cycle arrest. Of note, JI051 dose-dependently reduced cell growth of the human pancreatic cancer cell line MIA PaCa-2, and JI130 treatment significantly reduced tumor volume in a murine pancreatic tumor xenograft model. These results suggest a previously unrecognized role for PHB2 in the regulation of Hes1 and may inform potential strategies for managing pancreatic cancer.
Collapse
Affiliation(s)
- Amelie Perron
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011; Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Uji, Kyoto 611-0011
| | | | - Jun Iwata
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011
| | - Hiromi Shimojo
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Uji, Kyoto 611-0011; Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Junichiro Takaya
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011
| | - Kumiko Kobayashi
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Itaru Imayoshi
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8507, Japan
| | - Naasson M Mbenza
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011
| | - Mihoko Takenoya
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Uji, Kyoto 611-0011
| | - Ryoichiro Kageyama
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Uji, Kyoto 611-0011; Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Yuzo Kodama
- Department of Gastroenterology and Hepatology, Graduate School of Medicine.
| | - Motonari Uesugi
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011; Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Uji, Kyoto 611-0011.
| |
Collapse
|
16
|
Simutis FJ, Sanderson TP, Pilcher GD, Graziano MJ. Nonclinical Safety Assessment of the γ-Secretase Inhibitor Avagacestat. Toxicol Sci 2018. [DOI: 10.1093/toxsci/kfy048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Frank J Simutis
- Drug Safety Evaluation, Bristol-Myers Squibb Research and Development, New Brunswick, New Jersey 08903
| | - Thomas P Sanderson
- Drug Safety Evaluation, Bristol-Myers Squibb Research and Development, New Brunswick, New Jersey 08903
| | - Gary D Pilcher
- Drug Safety Evaluation, Bristol-Myers Squibb Research and Development, New Brunswick, New Jersey 08903
| | - Michael J Graziano
- Drug Safety Evaluation, Bristol-Myers Squibb Research and Development, New Brunswick, New Jersey 08903
| |
Collapse
|
17
|
Lovell MA, Lynn BC, Fister S, Bradley-Whitman M, Murphy MP, Beckett TL, Norris CM. A Novel Small Molecule Modulator of Amyloid Pathology. J Alzheimers Dis 2018; 53:273-87. [PMID: 27163808 DOI: 10.3233/jad-151160] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Because traditional approaches to drug development for Alzheimer's disease are becoming increasingly expensive and in many cases disappointingly unsuccessful, alternative approaches are required to shift the paradigm. Following leads from investigations of dihydropyridine calcium channel blockers, we observed unique properties from a class of functionalized naphthyridines and sought to develop these as novel therapeutics that minimize amyloid pathology without the adverse effects associated with current therapeutics. Our data show methyl 2,4-dimethyl-5-oxo-5,6-dihydrobenzo[c][2,7]naphthyridine-1-carboxylate (BNC-1) significantly decreases amyloid burden in a well-established mouse model of amyloid pathology through a unique mechanism mediated by Elk-1, a transcriptional repressor of presenilin-1. Additionally, BNC-1 treatment leads to increased levels of synaptophysin and synapsin, markers of synaptic integrity, but does not adversely impact presenilin-2 or processing of Notch-1, thus avoiding negative off target effects associated with pan-gamma secretase inhibition. Overall, our data show BNC-1 significantly decreases amyloid burden and improves markers of synaptic integrity in a well-established mouse model of amyloid deposition by promoting phosphorylation and activation of Elk-1, a transcriptional repressor of presenilin-1 but not presenilin-2. These data suggest BNC-1 might be a novel, disease-modifying therapeutic that will alter the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Mark A Lovell
- Department of Chemistry, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Bert C Lynn
- Department of Chemistry, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.,Universisty of Kentucky Mass Spectrometry Center, Lexington, KY, USA
| | - Shuling Fister
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | | | - M Paul Murphy
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.,Department of Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Tina L Beckett
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Christopher M Norris
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.,Department of Pharmacology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
18
|
Amini-Nik S, Yousuf Y, Jeschke MG. Scar management in burn injuries using drug delivery and molecular signaling: Current treatments and future directions. Adv Drug Deliv Rev 2018; 123:135-154. [PMID: 28757325 PMCID: PMC5742037 DOI: 10.1016/j.addr.2017.07.017] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022]
Abstract
In recent decades, there have been tremendous improvements in burn care that have allowed patients to survive severe burn injuries that were once fatal. However, a major limitation of burn care currently is the development of hypertrophic scars in approximately 70% of patients. This significantly decreases the quality of life for patients due to the physical and psychosocial symptoms associated with scarring. Current approaches to manage scarring include surgical techniques and non-surgical methods such as laser therapy, steroid injections, and compression therapy. These treatments are limited in their effectiveness and regularly fail to manage symptoms. As a result, the development of novel treatments that aim to improve outcomes and quality of life is imperative. Drug delivery that targets the molecular cascades of wound healing to attenuate or prevent hypertrophic scarring is a promising approach that has therapeutic potential. In this review, we discuss current treatments for scar management after burn injury, and how drug delivery targeting molecular signaling can lead to new therapeutic strategies.
Collapse
Affiliation(s)
- Saeid Amini-Nik
- Sunnybrook Research Institute, Toronto, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada.
| | - Yusef Yousuf
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada
| | - Marc G Jeschke
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada; Department of Immunology, University of Toronto, Toronto, Canada; Ross-Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Canada.
| |
Collapse
|
19
|
Zarone MR, Misso G, Grimaldi A, Zappavigna S, Russo M, Amler E, Di Martino MT, Amodio N, Tagliaferri P, Tassone P, Caraglia M. Evidence of novel miR-34a-based therapeutic approaches for multiple myeloma treatment. Sci Rep 2017; 7:17949. [PMID: 29263373 PMCID: PMC5738363 DOI: 10.1038/s41598-017-18186-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 12/01/2017] [Indexed: 02/07/2023] Open
Abstract
MiR-34a acts as tumor suppressor microRNA (miRNA) in several cancers, including multiple myeloma (MM), by controlling the expression of target proteins involved in cell cycle, differentiation and apoptosis. Here, we have investigated the combination between miR-34a and γ-secretase inhibitor (γSI), Sirtinol or zoledronic acid (ZOL) in order to enhance the inhibitory action of this miRNA on its canonical targets such as Notch1 and SIRT1, and on Ras/MAPK-dependent pathways. Our data demonstrate that miR-34a synthetic mimics significantly enhance the anti-tumor activity of all the above-mentioned anti-cancer agents in RPMI 8226 MM cells. We found that γSI enhanced miR-34a-dependent anti-tumor effects by activating the extrinsic apoptotic pathway which could overcome the cytoprotective autophagic mechanism. Moreover, the combination between miR-34a and γSI increased the cell surface calreticulin (CRT) expression, that is well known for triggering anti-tumor immunological response. The combination between miR-34a and Sirtinol induced the activation of an intrinsic apoptotic pathway along with increased surface expression of CRT. Regarding ZOL, we found a powerful growth inhibition after enforced miR-34a expression, which was not likely attributable to neither apoptosis nor autophagy modulation. Based on our data, the combination of miR-34a with other anti-cancer agents appears a promising anti-MM strategy deserving further investigation.
Collapse
Affiliation(s)
- Mayra Rachele Zarone
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Gabriella Misso
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Anna Grimaldi
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Silvia Zappavigna
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Margherita Russo
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Evzen Amler
- Institute of Biophysics, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
- Laboratory of Tissue Engineering, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Catanzaro, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy.
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
20
|
Notch inhibitors and their role in the treatment of triple negative breast cancer: promises and failures. Curr Opin Oncol 2017; 29:411-427. [DOI: 10.1097/cco.0000000000000406] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
21
|
Stem Cell Technology for (Epi)genetic Brain Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 978:443-475. [PMID: 28523560 DOI: 10.1007/978-3-319-53889-1_23] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite the enormous efforts of the scientific community over the years, effective therapeutics for many (epi)genetic brain disorders remain unidentified. The common and persistent failures to translate preclinical findings into clinical success are partially attributed to the limited efficiency of current disease models. Although animal and cellular models have substantially improved our knowledge of the pathological processes involved in these disorders, human brain research has generally been hampered by a lack of satisfactory humanized model systems. This, together with our incomplete knowledge of the multifactorial causes in the majority of these disorders, as well as a thorough understanding of associated (epi)genetic alterations, has been impeding progress in gaining more mechanistic insights from translational studies. Over the last years, however, stem cell technology has been offering an alternative approach to study and treat human brain disorders. Owing to this technology, we are now able to obtain a theoretically inexhaustible source of human neural cells and precursors in vitro that offer a platform for disease modeling and the establishment of therapeutic interventions. In addition to the potential to increase our general understanding of how (epi)genetic alterations contribute to the pathology of brain disorders, stem cells and derivatives allow for high-throughput drugs and toxicity testing, and provide a cell source for transplant therapies in regenerative medicine. In the current chapter, we will demonstrate the validity of human stem cell-based models and address the utility of other stem cell-based applications for several human brain disorders with multifactorial and (epi)genetic bases, including Parkinson's disease (PD), Alzheimer's disease (AD), fragile X syndrome (FXS), Angelman syndrome (AS), Prader-Willi syndrome (PWS), and Rett syndrome (RTT).
Collapse
|
22
|
Sogorb-Esteve A, García-Ayllón MS, Llansola M, Felipo V, Blennow K, Sáez-Valero J. Inhibition of γ-Secretase Leads to an Increase in Presenilin-1. Mol Neurobiol 2017; 55:5047-5058. [PMID: 28815510 PMCID: PMC5948247 DOI: 10.1007/s12035-017-0705-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 08/01/2017] [Indexed: 12/27/2022]
Abstract
γ-Secretase inhibitors (GSIs) are potential therapeutic agents for Alzheimer’s disease (AD); however, trials have proven disappointing. We addressed the possibility that γ-secretase inhibition can provoke a rebound effect, elevating the levels of the catalytic γ-secretase subunit, presenilin-1 (PS1). Acute treatment of SH-SY5Y cells with the GSI LY-374973 (N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester, DAPT) augments PS1, in parallel with increases in other γ-secretase subunits nicastrin, presenilin enhancer 2, and anterior pharynx-defective 1, yet with no increase in messenger RNA expression. Over-expression of the C-terminal fragment (CTF) of APP, C99, also triggered an increase in PS1. Similar increases in PS1 were evident in primary neurons treated repeatedly (4 days) with DAPT or with the GSI BMS-708163 (avagacestat). Likewise, rats examined after 21 days administered with avagacestat (40 mg/kg/day) had more brain PS1. Sustained γ-secretase inhibition did not exert a long-term effect on PS1 activity, evident through the decrease in CTFs of APP and ApoER2. Prolonged avagacestat treatment of rats produced a subtle impairment in anxiety-like behavior. The rebound increase in PS1 in response to GSIs must be taken into consideration for future drug development.
Collapse
Affiliation(s)
- Aitana Sogorb-Esteve
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Av. Ramón y Cajal s/n, 03550, Sant Joan d'Alacant, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sant Joan d'Alacant, Spain
| | - María-Salud García-Ayllón
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Av. Ramón y Cajal s/n, 03550, Sant Joan d'Alacant, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sant Joan d'Alacant, Spain. .,Unidad de Investigación, Hospital General Universitario de Elche, FISABIO, 03203, Elche, Spain.
| | - Marta Llansola
- Laboratory of Neurobiology, Fundación Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Fundación Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal Campus, Sweden
| | - Javier Sáez-Valero
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Av. Ramón y Cajal s/n, 03550, Sant Joan d'Alacant, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sant Joan d'Alacant, Spain.
| |
Collapse
|
23
|
The role of Notch signaling in gastric carcinoma: molecular pathogenesis and novel therapeutic targets. Oncotarget 2017; 8:53839-53853. [PMID: 28881855 PMCID: PMC5581154 DOI: 10.18632/oncotarget.17809] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/17/2017] [Indexed: 12/14/2022] Open
Abstract
Notch signaling, an evolutionarily conserved signaling cascade system, is involved in promoting the progression of different types of cancers. Within the past decades, the Notch signaling pathway has increasingly been shown to have a primary role in deciding the fate of cancer cells and cancer stem cells in the stomach. Most components of Notch signaling are strongly expressed at different levels in gastric carcinoma tissue samples and are associated with a considerable number of clinical parameters. Moreover, crosstalk signaling between the Notch pathway and the Wnt, Ras, and NF-κB pathways promotes the process of gastric carcinogenesis. Consequently, this increases proliferation and prevents apoptosis in gastric cancer cells, and it contributes to the induction of angiogenesis and accelerates the progression of the epithelial-to-mesenchymal transition. Although the Notch signaling pathway presents novel therapeutic targets for cancer therapeutic intervention, there is still a dearth of in-depth understanding of the molecular mechanisms of Notch signaling in gastric carcinoma. In this review, we summarize the landscape of the Notch signaling pathway and recent findings on Notch signaling in gastric cancer. Furthermore, advanced studies and clinical treatments targeting the Notch signaling pathway arediscussed.
Collapse
|
24
|
Mundinger TO, Taborsky GJ. Early sympathetic islet neuropathy in autoimmune diabetes: lessons learned and opportunities for investigation. Diabetologia 2016; 59:2058-67. [PMID: 27342407 PMCID: PMC6214182 DOI: 10.1007/s00125-016-4026-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 05/23/2016] [Indexed: 12/13/2022]
Abstract
This review outlines the current state of knowledge regarding a unique neural defect of the pancreatic islet in autoimmune diabetes, one that we have termed early sympathetic islet neuropathy (eSIN). We begin with the findings that a majority of islet sympathetic nerves are lost near the onset of type 1, but not type 2, diabetes and that this nerve loss is restricted to the islet. We discuss later work demonstrating that while the loss of islet sympathetic nerves and the loss of islet beta cells in type 1 diabetes both require infiltration of the islet by lymphocytes, their respective mechanisms of tissue destruction differ. Uniquely, eSIN requires the activation of a specific neurotrophin receptor and we propose two possible pathways for activation of this receptor during the immune attack on the islet. We also outline what is known about the functional consequences of eSIN, focusing on impairment of sympathetically mediated glucagon secretion and its application to the clinical problem of insulin-induced hypoglycaemia. Finally, we offer our view on the important remaining questions regarding this unique neural defect.
Collapse
Affiliation(s)
- Thomas O Mundinger
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, 98105, USA.
- Veterans Affairs Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA.
| | - Gerald J Taborsky
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, 98105, USA
- Veterans Affairs Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| |
Collapse
|
25
|
Soares HD, Gasior M, Toyn JH, Wang JS, Hong Q, Berisha F, Furlong MT, Raybon J, Lentz KA, Sweeney F, Zheng N, Akinsanya B, Berman RM, Thompson LA, Olson RE, Morrison J, Drexler DM, Macor JE, Albright CF, Ahlijanian MK, AbuTarif M. The γ-Secretase Modulator, BMS-932481, Modulates Aβ Peptides in the Plasma and Cerebrospinal Fluid of Healthy Volunteers. J Pharmacol Exp Ther 2016; 358:138-50. [PMID: 27189973 PMCID: PMC4931877 DOI: 10.1124/jpet.116.232256] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/05/2016] [Indexed: 11/22/2022] Open
Abstract
The pharmacokinetics, pharmacodynamics, safety, and tolerability of BMS-932481, a γ-secretase modulator (GSM), were tested in healthy young and elderly volunteers after single and multiple doses. BMS-932481 was orally absorbed, showed dose proportionality after a single dose administration, and had approximately 3-fold accumulation after multiple dosing. High-fat/caloric meals doubled the Cmax and area under the curve and prolonged Tmax by 1.5 hours. Consistent with the preclinical pharmacology of GSMs, BMS-932481 decreased cerebrospinal fluid (CSF) Aβ39, Aβ40, and Aβ42 while increasing Aβ37 and Aβ38, thereby providing evidence of γ-secretase enzyme modulation rather than inhibition. In plasma, reductions in Aβ40 and Aβ42 were observed with no change in total Aβ; in CSF, modest decreases in total Aβ were observed at higher dose levels. Increases in liver enzymes were observed at exposures associated with greater than 70% CSF Aβ42 lowering after multiple dosing. Although further development was halted due to an insufficient safety margin to test the hypothesis for efficacy of Aβ lowering in Alzheimer's disease, this study demonstrates that γ-secretase modulation is achievable in healthy human volunteers and supports further efforts to discover well tolerated GSMs for testing in Alzheimer's disease and other indications.
Collapse
Affiliation(s)
- Holly D Soares
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Maciej Gasior
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Jeremy H Toyn
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Jun-Sheng Wang
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Quan Hong
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Flora Berisha
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Michael T Furlong
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Joseph Raybon
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Kimberley A Lentz
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Francis Sweeney
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Naiyu Zheng
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Billy Akinsanya
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Robert M Berman
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Lorin A Thompson
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Richard E Olson
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - John Morrison
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Dieter M Drexler
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - John E Macor
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Charlie F Albright
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Michael K Ahlijanian
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Malaz AbuTarif
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| |
Collapse
|
26
|
Huang CK, Iwagami Y, Aihara A, Chung W, de la Monte S, Thomas JM, Olsen M, Carlson R, Yu T, Dong X, Wands J. Anti-Tumor Effects of Second Generation β-Hydroxylase Inhibitors on Cholangiocarcinoma Development and Progression. PLoS One 2016; 11:e0150336. [PMID: 26954680 PMCID: PMC4783022 DOI: 10.1371/journal.pone.0150336] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 02/11/2016] [Indexed: 12/19/2022] Open
Abstract
Cholangiocarcinoma (CCA) has a poor prognosis due to widespread intrahepatic spread. Aspartate β-hydroxylase (ASPH) is a transmembrane protein and catalyzes the hydroxylation of aspartyl and asparaginyl residues in calcium binding epidermal growth factor (cbEGF)-like domains of various proteins, including Notch receptors and ligands. ASPH is highly overexpressed (>95%) in human CCA tumors. We explored the molecular mechanisms by which ASPH mediated the CCA malignant phenotype and evaluated the potential of ASPH as a therapeutic target for CCA. The importance of expression and enzymatic activity of ASPH for CCA growth and progression was examined using shRNA "knockdown" and a mutant construct that reduced its catalytic activity. Second generation small molecule inhibitors (SMIs) of β-hydroxylase activity were developed and used to target ASPH in vitro and in vivo. Subcutaneous and intrahepatic xenograft rodent models were employed to determine anti-tumor effects on CCA growth and development. It was found that the enzymatic activity of ASPH was critical for mediating CCA progression, as well as inhibiting apoptosis. Mechanistically, ASPH overexpression promoted Notch activation and modulated CCA progression through a Notch1-dependent cyclin D1 pathway. Targeting ASPH with shRNAs or a SMI significantly suppressed CCA growth in vivo.
Collapse
Affiliation(s)
- Chiung-Kuei Huang
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Yoshifumi Iwagami
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Arihiro Aihara
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Waihong Chung
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Suzanne de la Monte
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - John-Michael Thomas
- Department of Medical Chemistry, College of Pharmacy Glendale, Midwestern University, Glendale, Arizona, United States of America
| | - Mark Olsen
- Department of Medical Chemistry, College of Pharmacy Glendale, Midwestern University, Glendale, Arizona, United States of America
| | - Rolf Carlson
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Tunan Yu
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Xiaoqun Dong
- Department of Biomedical and Pharmaceutical Science, College of Pharmacy, The University of Rhode Island, Pharmacy Building, 7 Greenhouse Road, Kingston, Rhode Island, United States of America
| | - Jack Wands
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
- * E-mail:
| |
Collapse
|
27
|
Calcium Channel Blockers, Progression to Dementia, and Effects on Amyloid Beta Peptide Production. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015. [PMID: 26221415 PMCID: PMC4499419 DOI: 10.1155/2015/787805] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Previous epidemiologic studies suggest that antihypertensive drugs may be protective against cognitive decline. To determine if subjects enrolled in the University of Kentucky longitudinal aging study who used antihypertensive drugs showed diminished progression to dementia, we used a 3-parameter logistic regression model to compare the rate of progression to dementia for subjects who used any of the five common categories of antihypertensive drugs to those with similar demographic characteristics but who did not use antihypertensives. Regression modeling showed that subjects who used calcium channel blockers (CCBs) but not the other classes of antihypertensives showed a significant decrease in the rate of progression to dementia. Significantly, use of CCBs ameliorated the negative effects of the presence of APOE-4 alleles on cognitive decline. To determine if CCBs could minimize amyloid beta peptide (Aβ1–42) production, H4 neuroglioma cultures transfected to overexpress APP were treated with various CCBs and Aβ1–42 levels and levels of proteins involved in Aβ production were quantified. Results show that treatment with nifedipine led to a significant decrease in levels of Aβ1–42, with no significant decrease in cell viability. Collectively, these data suggest that use of CCBs significantly diminishes the rate of progression to dementia and may minimize formation of Aβ1–42.
Collapse
|
28
|
Doody RS, Raman R, Sperling RA, Seimers E, Sethuraman G, Mohs R, Farlow M, Iwatsubo T, Vellas B, Sun X, Ernstrom K, Thomas RG, Aisen PS. Peripheral and central effects of γ-secretase inhibition by semagacestat in Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2015; 7:36. [PMID: 26064192 PMCID: PMC4461930 DOI: 10.1186/s13195-015-0121-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 03/16/2015] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The negative efficacy study examining the γ-secretase inhibitor semagacestat in mild to moderate Alzheimer's disease (AD) included a number of biomarkers of the disease as well as safety outcomes. We analyzed these data to explore relationships between drug exposure and pharmacodynamic effects and to examine the correlations among outcome measures. METHODS The study was a multicenter, randomized, placebo-controlled trial of two dose regimens of semagacestat and a placebo administered for 18 months to individuals with mild to moderate AD. Changes in measures of central and peripheral drug activity were compared between the three treatment groups using one-way analysis of variance. The relationship between changes in each of the outcome measures and measures of drug exposure and peripheral pharmacodynamic effect were assessed using Spearman's correlation coefficient. RESULTS Assignment to the active treatment arms was associated with reduction in plasma amyloid-β (Aβ) peptides, increase in ventricular volume, decrease in cerebrospinal fluid phosphorylated tau (p-tau) and several other laboratory measures and adverse event categories. Within the active arms, exposure to drug, as indicated by area under the concentration curve (AUC) of blood concentration, was associated with reduction in plasma Aβ peptides and a subset of laboratory changes and adverse event rates. Ventricular volume increase, right hippocampal volume loss and gastrointestinal symptoms were related to change in plasma Aβ peptide but not AUC, supporting a link to inhibition of γ-secretase cleavage of the amyloid precursor protein. Cognitive decline correlated with ventricular expansion and reduction in p-tau. CONCLUSION These findings may inform future studies of drugs targeting secretases involved in Aβ generation. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT00594568. Registered 11 January 2008.
Collapse
Affiliation(s)
- Rachelle S Doody
- Alzheimer's Disease and Memory Disorders center, Department of Neurology, Baylor College of Medicine, 1977 Butler Blvd, Suite E5.101 Houston, TX USA
| | - Rema Raman
- Division of Biostatistics and Bioinformatics, Department of Family and Preventive Medicine, Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, M/C 0949, La Jolla, CA 92093 USA ; Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, M/C 0949, La Jolla, CA, 92093 USA
| | - Reisa A Sperling
- Department of Neurology, Harvard Medical School, 220 Longwood Avenue, Goldenson Building, Room 420, Boston, MA 02115 USA
| | - Eric Seimers
- Eli Lilly & Company, Lilly Corporate Center, Indianapolis, IN 46285 USA
| | | | - Richard Mohs
- Eli Lilly & Company, Lilly Corporate Center, Indianapolis, IN 46285 USA
| | - Martin Farlow
- Department of Neurology, Indiana University, Indiana Alzheimer Disease Center, 355 W. 16th Street, Suite 4700, Indianapolis, IN 46202 USA
| | - Takeshi Iwatsubo
- Graduate School of Medicine, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8654 Japan
| | - Bruno Vellas
- Gerontopole UMR INSERM 1027, CHU, University of Toulouse, Toulouse, France
| | - Xiaoying Sun
- Biostatistics Research Center, Department of Family and Preventive Medicine, University of California San Diego, 9500 Gilman Drive, M/C 0949, La Jolla, CA 92093 USA
| | - Karin Ernstrom
- Biostatistics Research Center, Department of Family and Preventive Medicine, University of California San Diego, 9500 Gilman Drive, M/C 0949, La Jolla, CA 92093 USA
| | - Ronald G Thomas
- Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, M/C 0949, La Jolla, CA, 92093 USA ; Alzheimer's Disease Cooperative Study, Department of Family and Preventive Medicine, Department of Neurosciences, University of California San Diego, 9500 Gilman Drive M/C 0949, La Jolla, CA 92093 USA
| | - Paul S Aisen
- Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, M/C 0949, La Jolla, CA, 92093 USA
| | | |
Collapse
|
29
|
Zheng X, Liu D, Klärner FG, Schrader T, Bitan G, Bowers MT. Amyloid β-protein assembly: The effect of molecular tweezers CLR01 and CLR03. J Phys Chem B 2015; 119:4831-41. [PMID: 25751170 PMCID: PMC4415044 DOI: 10.1021/acs.jpcb.5b00692] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
![]()
The early oligomerization of amyloid
β-protein (Aβ)
has been shown to be an important event in the pathology of Alzheimer’s
disease (AD). Designing small molecule inhibitors targeting Aβ
oligomerization is one attractive and promising strategy for AD treatment.
Here we used ion mobility spectrometry coupled to mass spectrometry
(IMS-MS) to study the different effects of the molecular tweezers
CLR01 and CLR03 on Aβ self-assembly. CLR01 was found to bind
to Aβ directly and disrupt its early oligomerization. Moreover,
CLR01 remodeled the early oligomerization of Aβ42 by compacting
the structures of dimers and tetramers and as a consequence eliminated
higher-order oligomers. Unexpectedly, the negative-control derivative,
CLR03, which lacks the hydrophobic arms of the tweezer structure,
was found to facilitate early Aβ oligomerization. Our study
provides an example of IMS as a powerful tool to study and better
understand the interaction between small molecule modulators and Aβ
oligomerization, which is not attainable by other methods, and provides
important insights into therapeutic development of molecular tweezers
for AD treatment.
Collapse
Affiliation(s)
- Xueyun Zheng
- †Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Deyu Liu
- †Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Frank-Gerrit Klärner
- ‡Institute of Organic Chemistry, University of Duisburg-Essen, Essen 45117, Germany
| | - Thomas Schrader
- ‡Institute of Organic Chemistry, University of Duisburg-Essen, Essen 45117, Germany
| | - Gal Bitan
- §Department of Neurology, David Geffen School of Medicine, Brain Research Institute, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Michael T Bowers
- †Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
30
|
Lannfelt L, Möller C, Basun H, Osswald G, Sehlin D, Satlin A, Logovinsky V, Gellerfors P. Perspectives on future Alzheimer therapies: amyloid-β protofibrils - a new target for immunotherapy with BAN2401 in Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2014; 6:16. [PMID: 25031633 PMCID: PMC4054967 DOI: 10.1186/alzrt246] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The symptomatic drugs currently on the market for Alzheimer's disease (AD) have no effect on disease progression, and this creates a large unmet medical need. The type of drug that has developed most rapidly in the last decade is immunotherapy: vaccines and, especially, passive vaccination with monoclonal antibodies. Antibodies are attractive drugs as they can be made highly specific for their target and often with few side effects. Data from recent clinical AD trials indicate that a treatment effect by immunotherapy is possible, providing hope for a new generation of drugs. The first anti-amyloid-beta (anti-Aβ) vaccine developed by Elan, AN1792, was halted in phase 2 because of aseptic meningoencephalitis. However, in a follow-up study, patients with antibody response to the vaccine demonstrated reduced cognitive decline, supporting the hypothesis that Aβ immunotherapy may have clinically relevant effects. Bapineuzumab (Elan/Pfizer Inc./Johnson & Johnson), a monoclonal antibody targeting fibrillar Aβ, was stopped because the desired clinical effect was not seen. Solanezumab (Eli Lilly and Company) was developed to target soluble, monomeric Aβ. In two phase 3 studies, Solanezumab did not meet primary endpoints. When data from the two studies were pooled, a positive pattern emerged, revealing a significant slowing of cognitive decline in the subgroup of mild AD. The Arctic mutation has been shown to specifically increase the formation of soluble Aβ protofibrils, an Aβ species shown to be toxic to neurons and likely to be present in all cases of AD. A monoclonal antibody, mAb158, was developed to target Aβ protofibrils with high selectivity. It has at least a 1,000-fold higher selectivity for protofibrils as compared with monomers of Aβ, thus targeting the toxic species of the peptide. A humanized version of mAb158, BAN2401, has now entered a clinical phase 2b trial in a collaboration between BioArctic Neuroscience and Eisai without the safety concerns seen in previous phase 1 and 2a trials. Experiences from the field indicate the importance of initiating treatment early in the course of the disease and of enriching the trial population by improving the diagnostic accuracy. BAN2401 is a promising candidate for Aβ immunotherapy in early AD. Other encouraging efforts in immunotherapy as well as in the small-molecule field offer hope for new innovative therapies for AD in the future.
Collapse
Affiliation(s)
- Lars Lannfelt
- Department of Public Health/Geriatrics, Uppsala University, Dag Hammarskölds väg 14 B, 751 85 Uppsala, Sweden ; BioArctic Neuroscience AB, Warfvinges väg 35, 112 51 Stockholm, Sweden
| | - Christer Möller
- BioArctic Neuroscience AB, Warfvinges väg 35, 112 51 Stockholm, Sweden
| | - Hans Basun
- Department of Public Health/Geriatrics, Uppsala University, Dag Hammarskölds väg 14 B, 751 85 Uppsala, Sweden ; BioArctic Neuroscience AB, Warfvinges väg 35, 112 51 Stockholm, Sweden
| | - Gunilla Osswald
- BioArctic Neuroscience AB, Warfvinges väg 35, 112 51 Stockholm, Sweden
| | - Dag Sehlin
- Department of Public Health/Geriatrics, Uppsala University, Dag Hammarskölds väg 14 B, 751 85 Uppsala, Sweden
| | - Andrew Satlin
- Eisai, Inc, 100 Tice Boulevard, Woodcliff Lake, NJ 07677, USA
| | | | - Pär Gellerfors
- BioArctic Neuroscience AB, Warfvinges väg 35, 112 51 Stockholm, Sweden
| |
Collapse
|
31
|
Abstract
Current treatment options for Alzheimer's disease (AD) are limited to medications that reduce dementia symptoms. Given the rapidly ageing populations in most areas of the world, new therapeutic interventions for AD are urgently needed. In recent years, a number of drug candidates targeting the amyloid-ß (Aß) peptide have advanced into clinical trials; however, most have failed because of safety issues or lack of efficacy. The Aß peptide is central to the pathogenesis, and immunotherapy against Aß has attracted considerable interest. It offers the possibility to reach the target with highly specific drugs. Active immunization and passive immunization have been the most widely studied approaches to immunotherapy of AD. A favourable aspect of active immunization is the capacity for a small number of vaccinations to generate a prolonged antibody response. A potential disadvantage is the variability in the antibody response across patients. The potential advantages of passive immunotherapy include the reproducible delivery of a known amount of therapeutic antibodies to the patient and rapid clearance of those antibodies if side effects develop. A disadvantage is the requirement for repeated infusions of antibodies over time. After more than a decade of research, anti-amyloid immunotherapy remains one of the most promising emerging strategies for developing disease-modifying treatments for AD. In this review, we examine the presently ongoing Aß-directed immunotherapies that have passed clinical development Phase IIa.
Collapse
Affiliation(s)
- L Lannfelt
- Department of Public Health/Geriatrics, Uppsala University, Uppsala, Sweden
| | | | | |
Collapse
|
32
|
Léger GC, Massoud F. Novel disease-modifying therapeutics for the treatment of Alzheimer’s disease. Expert Rev Clin Pharmacol 2014; 6:423-42. [DOI: 10.1586/17512433.2013.811237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
33
|
Mikulca JA, Nguyen V, Gajdosik DA, Teklu SG, Giunta EA, Lessa EA, Tran CH, Terak EC, Raffa RB. Potential novel targets for Alzheimer pharmacotherapy: II. Update on secretase inhibitors and related approaches. J Clin Pharm Ther 2013; 39:25-37. [PMID: 24313554 DOI: 10.1111/jcpt.12112] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 10/29/2013] [Indexed: 11/26/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE The prevailing theory regarding Alzheimer disease (AD) is that insoluble amyloid β-peptide (Aβ) plays a critical role in the cortical plaques characteristic of the disease. Because Aβ is formed from the sequential splicing of amyloid precursor protein (APP) catalysed by 'secretase' enzymes (α, β and γ), clinical trials of secretase inhibitors will either result in beneficial pharmacotherapy or, if negative, cast doubt on the role of Aβ in AD. With recent clinical trial failures, is the Aβ theory wrong? METHODS Literature searches were conducted on the topics of secretases and clinical trials, including PubMed searches, United States clinical trials directory, pharmaceutical company websites and news reports. The information was collected and evaluated for relevance and quality. RESULTS AND DISCUSSION Several direct-acting (e.g. CTS-21166, LY2811376) and indirect-acting (e.g. ACI-91) β-secretase inhibitors and several γ-secretase inhibitors (e.g. avagacestat, JNJ-40418677 and semagacestat) have not fared well in early clinical trials due to the lack of efficacy or concerns over possible serious side effects. WHAT IS NEW AND CONCLUSION The failures of secretase inhibitors in clinical trials appear to bring into question the long-hypothesized association between AD and Aβ production. However, the disease might have been too advanced in these patients to benefit from this type of therapy (mainly preventive). Secretase inhibitors are still being studied, along with new diagnostic tools, with the hope of testing patients earlier, that is, with less advanced disease. If these trials also fail, the prevailing view of the role of Aβ in AD will truly be in doubt.
Collapse
Affiliation(s)
- J A Mikulca
- Temple University School of Pharmacy, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Zhou W, Wang G, Guo S. Regulation of angiogenesis via Notch signaling in breast cancer and cancer stem cells. Biochim Biophys Acta Rev Cancer 2013; 1836:304-20. [PMID: 24183943 DOI: 10.1016/j.bbcan.2013.10.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 10/14/2013] [Accepted: 10/18/2013] [Indexed: 02/07/2023]
Abstract
Breast cancer angiogenesis is elicited and regulated by a number of factors including the Notch signaling. Notch receptors and ligands are expressed in breast cancer cells as well as in the stromal compartment and have been implicated in carcinogenesis. Signals exchanged between neighboring cells through the Notch pathway can amplify and consolidate molecular differences, which eventually dictate cell fates. Notch signaling and its crosstalk with many signaling pathways play an important role in breast cancer cell growth, migration, invasion, metastasis and angiogenesis, as well as cancer stem cell (CSC) self-renewal. Therefore, significant attention has been paid in recent years toward the development of clinically useful antagonists of Notch signaling. Better understanding of the structure, function and regulation of Notch intracellular signaling pathways, as well as its complex crosstalk with other oncogenic signals in breast cancer cells will be essential to ensure rational design and application of new combinatory therapeutic strategies. Novel opportunities have emerged from the discovery of Notch crosstalk with inflammatory and angiogenic cytokines and their links to CSCs. Combinatory treatments with drugs designed to prevent Notch oncogenic signal crosstalk may be advantageous over λ secretase inhibitors (GSIs) alone. In this review, we focus on the more recent advancements in our knowledge of aberrant Notch signaling contributing to breast cancer angiogenesis, as well as its crosstalk with other factors contributing to angiogenesis and CSCs.
Collapse
Affiliation(s)
- Weiqiang Zhou
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146 North Huanghe St, Huanggu Dis, Shenyang City, Liaoning Pro 110034, PR China.
| | | | | |
Collapse
|
35
|
Gassman A, Hao LT, Bhoite L, Bradford CL, Chien CB, Beattie CE, Manfredi JP. Small molecule suppressors of Drosophila kinesin deficiency rescue motor axon development in a zebrafish model of spinal muscular atrophy. PLoS One 2013; 8:e74325. [PMID: 24023935 PMCID: PMC3762770 DOI: 10.1371/journal.pone.0074325] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 07/31/2013] [Indexed: 12/15/2022] Open
Abstract
Proximal spinal muscular atrophy (SMA) is the most common inherited motor neuropathy and the leading hereditary cause of infant mortality. Currently there is no effective treatment for the disease, reflecting a need for pharmacologic interventions that restore performance of dysfunctional motor neurons or suppress the consequences of their dysfunction. In a series of assays relevant to motor neuron biology, we explored the activities of a collection of tetrahydroindoles that were reported to alter the metabolism of amyloid precursor protein (APP). In Drosophila larvae the compounds suppressed aberrant larval locomotion due to mutations in the Khc and Klc genes, which respectively encode the heavy and light chains of kinesin-1. A representative compound of this class also suppressed the appearance of axonal swellings (alternatively termed axonal spheroids or neuritic beads) in the segmental nerves of the kinesin-deficient Drosophila larvae. Given the importance of kinesin-dependent transport for extension and maintenance of axons and their growth cones, three members of the class were tested for neurotrophic effects on isolated rat spinal motor neurons. Each compound stimulated neurite outgrowth. In addition, consistent with SMA being an axonopathy of motor neurons, the three axonotrophic compounds rescued motor axon development in a zebrafish model of SMA. The results introduce a collection of small molecules as pharmacologic suppressors of SMA-associated phenotypes and nominate specific members of the collection for development as candidate SMA therapeutics. More generally, the results reinforce the perception of SMA as an axonopathy and suggest novel approaches to treating the disease.
Collapse
Affiliation(s)
- Andrew Gassman
- Sera Prognostics, Inc., Salt Lake City, Utah, United States of America
| | - Le T. Hao
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, United States of America
| | - Leena Bhoite
- Technology Commercialization Office, University of Utah, Salt Lake City, Utah, United States of America
| | - Chad L. Bradford
- Sera Prognostics, Inc., Salt Lake City, Utah, United States of America
| | - Chi-Bin Chien
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
| | - Christine E. Beattie
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, United States of America
| | - John P. Manfredi
- Sfida BioLogic, Inc., Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
36
|
Abstract
The numerous processes involved in the etiology of breast cancer such as cell survival, metabolism, proliferation, differentiation, and angiogenesis are currently being elucidated. However, underlying mechanisms that drive breast cancer progression and drug resistance are still poorly understood. As we discuss here in detail, the Notch signaling pathway is an important regulatory component of normal breast development, cell fate of normal breast stem cells, and proliferation and survival of breast cancer initiating cells. Notch exerts a wide range of critical effects through a canonical pathway where it is expressed as a type I membrane precursor heterodimer followed by at least two subsequent cleavages induced by ligand engagement to ultimately release an intracellular form to function as a transcriptional activator. Notch and its ligands are overexpressed in breast cancer, and one method of effectively blocking Notch activity is preventing its cleavage at the cell surface with γ-secretase inhibitors. In the context of Notch signaling, the application of clinically relevant anti-Notch drugs in treatment regimens may contribute to novel therapeutic interventions and promote more effective clinical response in women with breast cancer.
Collapse
Affiliation(s)
- Roma Olsauskas-Kuprys
- The Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Chicago, Chicago, IL, USA
| | | | | |
Collapse
|
37
|
Portanova P, Notaro A, Pellerito O, Sabella S, Giuliano M, Calvaruso G. Notch inhibition restores TRAIL-mediated apoptosis via AP1-dependent upregulation of DR4 and DR5 TRAIL receptors in MDA-MB-231 breast cancer cells. Int J Oncol 2013; 43:121-30. [PMID: 23686163 DOI: 10.3892/ijo.2013.1945] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 03/14/2013] [Indexed: 11/06/2022] Open
Abstract
Notch is a family of transmembrane receptors whose activation through proteolytic cleavage by γ-secretase targets genes which participate in cell development, differentiation and tumorigenesis. Notch signaling is constitutively activated in various cancers, including breast cancer and its upregulation is usually related with poor clinical outcomes. Therefore, targeting Notch signaling with γ-secretase inhibitors (GSIs) is considered a promising strategy for cancer treatment. We report that the γ-secretase inhibitor-I (GSI-I) sensitizes human breast cancer cells to apoptosis mediated by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). The antiproliferative GSI-I/TRAIL synergism was stronger in ER-negative MDA-MB-231 breast cancer cells compared with ER-positive MCF-7 cells. In MDA-MB-231 cells, GSI-I treatment induced upregulation of DR4 and DR5 TRAIL receptors. This effect seemed to be related to the activation of the transcription factor AP1 that was a consequence of Notch inhibition, as demonstrated by Notch-1 silencing experiments. Combined treatment induced loss of the mitochondrial transmembrane potential and activation of caspases. GSI-I alone and/or GSI-I/TRAIL combination also induced a significant decrease in the levels of some survival factors (survivin, c-IAP-2, Bcl-xL, BimEL and pAKT) and upregulation of pro-apoptotic factors BimL, BimS and Noxa, enhancing the cytotoxic potential of the two drugs. Taken together, these results indicate for the first time that GSI-I/TRAIL combination could represent a novel and potentially effective tool for breast cancer treatment.
Collapse
Affiliation(s)
- Patrizia Portanova
- Dipartimento di Medicina traslazionale, Università del Piemonte Orientale, Novara, Italy
| | | | | | | | | | | |
Collapse
|
38
|
In Vivo Characterization of a Novel γ-Secretase Inhibitor SCH 697466 in Rodents and Investigation of Strategies for Managing Notch-Related Side Effects. Int J Alzheimers Dis 2013; 2013:823528. [PMID: 23573456 PMCID: PMC3612465 DOI: 10.1155/2013/823528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 11/27/2012] [Indexed: 11/17/2022] Open
Abstract
Substantial evidence implicates β-amyloid (Aβ) peptides in the etiology of Alzheimer's disease (AD). Aβ is produced by the proteolytic cleavage of the amyloid precursor protein by β- and γ-secretase suggesting that γ-secretase inhibition may provide therapeutic benefit for AD. Although many γ-secretase inhibitors have been shown to be potent at lowering Aβ, some have also been shown to have side effects following repeated administration. All of these side effects can be attributed to altered Notch signaling, another γ-secretase substrate. Here we describe the in vivo characterization of the novel γ-secretase inhibitor SCH 697466 in rodents. Although SCH 697466 was effective at lowering Aβ, Notch-related side effects in the intestine and thymus were observed following subchronic administration at doses that provided sustained and complete lowering of Aβ. However, additional studies revealed that both partial but sustained lowering of Aβand complete but less sustained lowering of Aβ were successful approaches for managing Notch-related side effects. Further, changes in several Notch-related biomarkers paralleled the side effect observations. Taken together, these studies demonstrated that, by carefully varying the extent and duration of Aβ lowering by γ-secretase inhibitors, it is possible to obtain robust and sustained lowering of Aβ without evidence of Notch-related side effects.
Collapse
|
39
|
Albright CF, Dockens RC, Meredith JE, Olson RE, Slemmon R, Lentz KA, Wang JS, Denton RR, Pilcher G, Rhyne PW, Raybon JJ, Barten DM, Burton C, Toyn JH, Sankaranarayanan S, Polson C, Guss V, White R, Simutis F, Sanderson T, Gillman KW, Starrett JE, Bronson J, Sverdlov O, Huang SP, Castaneda L, Feldman H, Coric V, Zaczek R, Macor JE, Houston J, Berman RM, Tong G. Pharmacodynamics of selective inhibition of γ-secretase by avagacestat. J Pharmacol Exp Ther 2013; 344:686-95. [PMID: 23275065 DOI: 10.1124/jpet.112.199356] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A hallmark of Alzheimer's disease (AD) pathology is the accumulation of brain amyloid β-peptide (Aβ), generated by γ-secretase-mediated cleavage of the amyloid precursor protein (APP). Therefore, γ-secretase inhibitors (GSIs) may lower brain Aβ and offer a potential new approach to treat AD. As γ-secretase also cleaves Notch proteins, GSIs can have undesirable effects due to interference with Notch signaling. Avagacestat (BMS-708163) is a GSI developed for selective inhibition of APP over Notch cleavage. Avagacestat inhibition of APP and Notch cleavage was evaluated in cell culture by measuring levels of Aβ and human Notch proteins. In rats, dogs, and humans, selectivity was evaluated by measuring plasma blood concentrations in relation to effects on cerebrospinal fluid (CSF) Aβ levels and Notch-related toxicities. Measurements of Notch-related toxicity included goblet cell metaplasia in the gut, marginal-zone depletion in the spleen, reductions in B cells, and changes in expression of the Notch-regulated hairy and enhancer of split homolog-1 from blood cells. In rats and dogs, acute administration of avagacestat robustly reduced CSF Aβ40 and Aβ42 levels similarly. Chronic administration in rats and dogs, and 28-day, single- and multiple-ascending-dose administration in healthy human subjects caused similar exposure-dependent reductions in CSF Aβ40. Consistent with the 137-fold selectivity measured in cell culture, we identified doses of avagacestat that reduce CSF Aβ levels without causing Notch-related toxicities. Our results demonstrate the selectivity of avagacestat for APP over Notch cleavage, supporting further evaluation of avagacestat for AD therapy.
Collapse
Affiliation(s)
- Charles F Albright
- Research and Development, Bristol-Myers Squibb, Wallingford, Connecticut 06492, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
GUO JIANLI, HE LEI, YUAN PING, WANG PENG, LU YANJUN, TONG FANGLI, WANG YU, YIN YANHUA, TIAN JUN, SUN JUN. ADAM10 overexpression in human non-small cell lung cancer correlates with cell migration and invasion through the activation of the Notch1 signaling pathway. Oncol Rep 2012; 28:1709-18. [DOI: 10.3892/or.2012.2003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 08/06/2012] [Indexed: 11/06/2022] Open
|
41
|
Developmental pathways in breast cancer and breast tumor-initiating cells: Therapeutic implications. Cancer Lett 2012; 317:115-26. [DOI: 10.1016/j.canlet.2011.11.028] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 11/20/2011] [Indexed: 12/13/2022]
|
42
|
Wells EM, Rao AAN, Scafidi J, Packer RJ. Neurotoxicity of biologically targeted agents in pediatric cancer trials. Pediatr Neurol 2012; 46:212-21. [PMID: 22490765 PMCID: PMC3626408 DOI: 10.1016/j.pediatrneurol.2012.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 02/10/2012] [Indexed: 02/07/2023]
Abstract
Biologically targeted agents offer the promise of delivering specific anticancer effects while limiting damage to healthy tissue, including the central and peripheral nervous systems. During the past 5-10 years, these agents were examined in preclinical and adult clinical trials, and are used with increasing frequency in children with cancer. This review evaluates current knowledge about neurotoxicity from biologically targeted anticancer agents, particularly those in pediatric clinical trials. For each drug, neurotoxicity data are reviewed in adult (particularly studies of brain tumors) and pediatric studies when available. Overall, these agents are well tolerated, with few serious neurotoxic effects. Data from younger patients are limited, and more neurotoxicity may occur in the pediatric population because these agents target pathways that control not only tumorigenesis but also neural maturation. Further investigation is needed into long-term neurologic effects, particularly in children.
Collapse
Affiliation(s)
- Elizabeth M. Wells
- Brain Tumor Institute, Children's National Medical Center, Washington, DC
- Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, DC
- Department of Neurology and Pediatrics, George Washington University, Washington, DC
| | - Amulya A. Nageswara Rao
- Brain Tumor Institute, Children's National Medical Center, Washington, DC
- Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, DC
- Department of Neurology and Pediatrics, George Washington University, Washington, DC
- Division of Pediatric Hematology/Oncology, Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Joseph Scafidi
- Brain Tumor Institute, Children's National Medical Center, Washington, DC
- Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, DC
- Department of Neurology and Pediatrics, George Washington University, Washington, DC
| | - Roger J. Packer
- Brain Tumor Institute, Children's National Medical Center, Washington, DC
- Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, DC
- Department of Neurology and Pediatrics, George Washington University, Washington, DC
- Communications should be addressed to: Dr. Packer; Department of Neurology; Children's National Medical Center; 111 Michigan Avenue NW; Washington, DC 20010.
| |
Collapse
|
43
|
Abstract
Regulated intramembrane proteolysis (RIP) is a highly conserved signaling paradigm whereby membrane-bound signaling proteins are cleaved in their transmembrane region and then released into the cytoplasm to act as signaling molecules. In most if not all cases intramembrane cleavage is preceded and regulated by a membrane proximal cleavage step called 'ectodomain shedding'. Here we will review the role of ectodomain shedding in RIP of the NOTCH signaling pathway, a highly conserved cell-cell communication pathway that mediates cell fate decisions during development and in adult tissues.
Collapse
|
44
|
Targeting Notch signaling for cancer therapeutic intervention. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2012; 65:191-234. [PMID: 22959027 DOI: 10.1016/b978-0-12-397927-8.00007-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Notch signaling pathway is an evolutionarily conserved, intercellular signaling cascade. The Notch proteins are single-pass receptors that are activated upon interaction with the Delta (or Delta-like) and Jagged/Serrate families of membrane-bound ligands. Association of ligand-receptor leads to proteolytic cleavages that liberate the Notch intracellular domain (NICD) from the plasma membrane. The NICD translocates to the nucleus, where it forms a complex with the DNA-binding protein CSL, displacing a histone deacetylase (HDAc)-corepressor (CoR) complex from CSL. Components of a transcriptional complex, such as MAML1 and histone acetyltransferases (HATs), are recruited to the NICD-CSL complex, leading to the transcriptional activation of Notch target genes. The Notch signaling pathway plays a critical role in cell fate decision, tissue patterning, morphogenesis, and is hence regarded as a developmental pathway. However, if this pathway goes awry, it contributes to cellular transformation and tumorigenesis. There is mounting evidence that this pathway is dysregulated in a variety of malignancies, and can behave as either an oncogene or a tumor suppressor depending upon cell context. This chapter highlights the current evidence for aberration of the Notch signaling pathway in a wide range of tumors from hematological cancers, such as leukemia and lymphoma, through to lung, skin, breast, pancreas, colon, prostate, ovarian, brain, and liver tumors. It proposes that the Notch signaling pathway may represent novel target for cancer therapeutic intervention.
Collapse
|
45
|
Reedijk M. Notch Signaling and Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 727:241-57. [DOI: 10.1007/978-1-4614-0899-4_18] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Kukar TL, Ladd TB, Robertson P, Pintchovski SA, Moore B, Bann MA, Ren Z, Jansen-West K, Malphrus K, Eggert S, Maruyama H, Cottrell BA, Das P, Basi GS, Koo EH, Golde TE. Lysine 624 of the amyloid precursor protein (APP) is a critical determinant of amyloid β peptide length: support for a sequential model of γ-secretase intramembrane proteolysis and regulation by the amyloid β precursor protein (APP) juxtamembrane region. J Biol Chem 2011; 286:39804-12. [PMID: 21868378 PMCID: PMC3220543 DOI: 10.1074/jbc.m111.274696] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 08/16/2011] [Indexed: 11/06/2022] Open
Abstract
γ-Secretase is a multiprotein intramembrane cleaving aspartyl protease (I-CLiP) that catalyzes the final cleavage of the amyloid β precursor protein (APP) to release the amyloid β peptide (Aβ). Aβ is the primary component of senile plaques in Alzheimer's disease (AD), and its mechanism of production has been studied intensely. γ-Secretase executes multiple cleavages within the transmembrane domain of APP, with cleavages producing Aβ and the APP intracellular domain (AICD), referred to as γ and ε, respectively. The heterogeneous nature of the γ cleavage that produces various Aβ peptides is highly relevant to AD, as increased production of Aβ 1-42 is genetically and biochemically linked to the development of AD. We have identified an amino acid in the juxtamembrane region of APP, lysine 624, on the basis of APP695 numbering (position 28 relative to Aβ) that plays a critical role in determining the final length of Aβ peptides released by γ-secretase. Mutation of this lysine to alanine (K28A) shifts the primary site of γ-secretase cleavage from 1-40 to 1-33 without significant changes to ε cleavage. These results further support a model where ε cleavage occurs first, followed by sequential proteolysis of the remaining transmembrane fragment, but extend these observations by demonstrating that charged residues at the luminal boundary of the APP transmembrane domain limit processivity of γ-secretase.
Collapse
Affiliation(s)
- Thomas L Kukar
- Emory University, School of Medicine, Department of Pharmacology, Center for Neurodegenerative Disease, Atlanta, Georgia 30322, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Speiser J, Foreman K, Drinka E, Godellas C, Perez C, Salhadar A, Erşahin Ç, Rajan P. Notch-1 and Notch-4 biomarker expression in triple-negative breast cancer. Int J Surg Pathol 2011; 20:139-45. [PMID: 22084425 DOI: 10.1177/1066896911427035] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Triple-negative breast cancer (TNBC) demonstrates lack of expression of hormone receptors and human epidermal growth factor receptor. However, there is no targeted therapy for TNBC. The authors analyzed 29 TNBC cases for Notch-1 and Notch-4 biomarker expression and subcellular location, Ki67 proliferation rate, and relevant clinical/survival data. Results demonstrated an unfavorable Ki67 rate in 90% of cases, Notch-1 expression in tumor and endothelial cells in 100% of cases, and Notch-4 expression in tumor cells in 73% of cases and endothelial cells in 100% of cases. Additionally, subcellular localization of Notch-1 and Notch-4 was predominantly nuclear and cytoplasmic. In conclusion, (a) the majority of TNBCs are high-grade infiltrating ductal carcinomas with high Ki67 proliferation rate and (b) both Notch-1 and Notch-4 receptors are overexpressed in tumor and vascular endothelial cells with subcellular localization different from that of hormone-positive breast cancer. Targeting Notch signaling with gamma secretase inhibitors should to be explored to further improve the survival rate of TNBC patients.
Collapse
Affiliation(s)
- Jodi Speiser
- Department of Pathology, Loyola University Medical Center, Maywood, IL 60153, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Stepan AF, Karki K, McDonald WS, Dorff PH, Dutra JK, DiRico KJ, Won A, Subramanyam C, Efremov IV, O’Donnell CJ, Nolan CE, Becker SL, Pustilnik LR, Sneed B, Sun H, Lu Y, Robshaw AE, Riddell D, O'Sullivan TJ, Sibley E, Capetta S, Atchison K, Hallgren AJ, Miller E, Wood A, Obach RS. Metabolism-Directed Design of Oxetane-Containing Arylsulfonamide Derivatives as γ-Secretase Inhibitors. J Med Chem 2011; 54:7772-83. [DOI: 10.1021/jm200893p] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Antonia F. Stepan
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kapil Karki
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - W. Scott McDonald
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Peter H. Dorff
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jason K. Dutra
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kenneth J. DiRico
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Annie Won
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Chakrapani Subramanyam
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ivan V. Efremov
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Christopher J. O’Donnell
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Charles E. Nolan
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Stacey L. Becker
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Leslie R. Pustilnik
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Blossom Sneed
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Hao Sun
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Yasong Lu
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ashley E. Robshaw
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - David Riddell
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Theresa J. O'Sullivan
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Evelyn Sibley
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Steven Capetta
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kevin Atchison
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Andrew J. Hallgren
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Emily Miller
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Anthony Wood
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - R. Scott Obach
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
49
|
Xiang J, Ouyang Y, Cui Y, Lin F, Ren J, Long M, Chen X, Wei J, Zhang H, Zhang H. Silencing of Notch3 Using shRNA driven by survivin promoter inhibits growth and promotes apoptosis of human T-cell acute lymphoblastic leukemia cells. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2011; 12:59-65. [PMID: 21940234 DOI: 10.1016/j.clml.2011.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 07/15/2011] [Accepted: 07/15/2011] [Indexed: 10/17/2022]
Abstract
BACKGROUND As a highly conserved system, the activation of the Notch pathway has been implicated in the tumorigenesis of various hematologic diseases, including leukemias, lymphomas, and multiple myeloma. The Notch3 receptor is frequently expressed in T-cell acute lymphoblastic leukemia (T-ALL). METHODS To explore its possibility as a therapeutic target for T-ALL, we investigated the effect of Notch3 silencing on Jurkat and SupT1 cells using a novel tumor-specific short hairpin RNA (shRNA) driven by survivin promoters. RESULTS We found that downregulated expression of Notch3 correlated with significant apoptosis and inhibition of proliferation. CONCLUSION These facts suggest that downregulating expression of Notch3 could attenuate the Notch signaling activity in T-ALL. All these results indicate that inhibition of Notch3 expression can result in potent antitumor activity in T-ALL.
Collapse
Affiliation(s)
- Jie Xiang
- Department of Clinical Laboratory and Research Center, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ercan C, van Diest PJ, Vooijs M. Mammary development and breast cancer: the role of stem cells. Curr Mol Med 2011; 11:270-85. [PMID: 21506923 DOI: 10.2174/156652411795678007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 02/14/2011] [Indexed: 12/15/2022]
Abstract
The mammary gland is a highly regenerative organ that can undergo multiple cycles of proliferation, lactation and involution, a process controlled by stem cells. The last decade much progress has been made in the identification of signaling pathways that function in these stem cells to control self-renewal, lineage commitment and epithelial differentiation in the normal mammary gland. The same signaling pathways that control physiological mammary development and homeostasis are also often found deregulated in breast cancer. Here we provide an overview on the functional and molecular identification of mammary stem cells in the context of both normal breast development and breast cancer. We discuss the contribution of some key signaling pathways with an emphasis on Notch receptor signaling, a cell fate determination pathway often deregulated in breast cancer. A further understanding of the biological roles of the Notch pathway in mammary stem cell behavior and carcinogenesis might be relevant for the development of future therapies.
Collapse
Affiliation(s)
- C Ercan
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | | | | |
Collapse
|