1
|
Young OJ, Dembele H, Rajwar A, Kwon IC, Ryu JH, Shih WM, Zeng YC. Cargo quantification of functionalized DNA origami for therapeutic application. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609963. [PMID: 39253502 PMCID: PMC11383041 DOI: 10.1101/2024.08.27.609963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
In recent years, notable advances in nanotechnology-based drug delivery have emerged. A particularly promising platform in this field is DNA origami-based nanoparticles, which offer highly programmable surfaces, providing precise control over the nanoscale spacing and stoichiometry of various cargo. These versatile particles are finding diverse applications ranging from basic molecular biology to diagnostics and therapeutics. This growing interest creates the need for effective methods to quantify cargo on DNA origami nanoparticles. Our study consolidates several previously validated methods focusing on gel-based and fluorescence-based techniques, including multiplexed quantification of protein, peptide, and nucleic acid cargo on these nanoparticles. This work may serve as a valuable resource for groups researchers keen on utilizing DNA origami-based nanoparticles in therapeutic applications.
Collapse
Affiliation(s)
- Olivia J. Young
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard-Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Hawa Dembele
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Anjali Rajwar
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Ick Chan Kwon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Ju Hee Ryu
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - William M. Shih
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yang C. Zeng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
2
|
Alshubramy MA, Alotaibi FS, Alkahtani HM, Alamry KA, Hussein MA. C3-Symmetric ligands in drug design: An overview of the challenges and opportunities ahead. Bioorg Med Chem Lett 2024; 103:129702. [PMID: 38490620 DOI: 10.1016/j.bmcl.2024.129702] [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: 01/02/2024] [Revised: 02/10/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
C3-symmetry is a type of star-shaped molecule consisting of a central core and three symmetrically attached chains. These molecules are used in drug discovery due to their unique three-fold rotational symmetry, which allows for specific binding interactions and improved molecular recognition. In this text, we provide an overview of synthetic approaches with C3-symmetry as a pharmaceutical tool: progress, challenges, and opportunities. C3-symmetric ligands offer both challenges and opportunities in drug design. Their unique symmetry can enhance binding interactions, but careful consideration of rigidity, synthetic complexity, and target compatibility is crucial. Further research and advancements in synthetic methods and modeling tools will likely drive their exploration in drug discovery, leading to the discovery of potent C3-symmetric ligands.
Collapse
Affiliation(s)
- Maha A Alshubramy
- Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Faez S Alotaibi
- Department of Chemistry, College of Science, Qassim University Buraidah 51452, Saudi Arabia
| | - Hamad M Alkahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Khalid A Alamry
- Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mahmoud A Hussein
- Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Chemistry Department, Faculty of Science, Assiut University, Assiut 71516, Egypt.
| |
Collapse
|
3
|
Mamuti M, Wang Y, Zhao YD, Wang JQ, Wang J, Fan YL, Xiao WY, Hou DY, Yang J, Zheng R, An HW, Wang H. A Polyvalent Peptide CD40 Nanoagonist for Targeted Modulation of Dendritic Cells and Amplified Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109432. [PMID: 35426184 DOI: 10.1002/adma.202109432] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/06/2022] [Indexed: 06/14/2023]
Abstract
Targeted immunomodulation through biomolecule-based nanostructures, especially to dendritic cells (DCs), holds great promise for effective cancer therapy. However, construction of high-performance agonist by mimicking natural ligand to activate immune cell signaling is a great challenge so far. Here, a peptide-based nanoagonist toward CD40 (PVA-CD40) with preorganized interfacial topological structure that activates lymph node DCs efficiently and persistently, achieving amplified immune therapeutic efficacy is described. The on-site fabrication of PVA-CD40 is realized through the click conjugation of two functional peptides including the "CD40 anchoring arm" and the "assembly-driving motor." The resultant polyvalent interface rapidly triggers the receptor oligomerization and downstream signaling. Strikingly, one shot administration of PVA-CD40 elicits maturation period of DCs up to 2.3-fold comparing to that of CD40 antibody. Finally, combining the PVA-CD40 with anti-PD-1 antibody results in subsequent inhibition of tumor growth in both B16F10 and 4T1 mice tumor models with survival rate up to 37%, while none of the mice survives in the clinically relevant CD40 mAb and anti-PD-1 combination-treated group. It is envisioned that the fabrication of antibody-like superstructures in vivo provides an efficient platform for modulating the duration of immune response to achieve optimal therapeutic efficacy.
Collapse
Affiliation(s)
- Muhetaerjiang Mamuti
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yi Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yong-Dan Zhao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jia-Qi Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Jie Wang
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yan-Lei Fan
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Wu-Yi Xiao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Da-Yong Hou
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Jia Yang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Rui Zheng
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hong-Wei An
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
4
|
Somoza M, Bertelli A, Pratto CA, Verdun RE, Campetella O, Mucci J. Trypanosoma cruzi Induces B Cells That Regulate the CD4 + T Cell Response. Front Cell Infect Microbiol 2022; 11:789373. [PMID: 35071041 PMCID: PMC8766854 DOI: 10.3389/fcimb.2021.789373] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022] Open
Abstract
Trypanosoma cruzi infection induces a polyclonal B cell proliferative response characterized by maturation to plasma cells, excessive generation of germinal centers, and secretion of parasite-unrelated antibodies. Although traditionally reduced to the humoral response, several infectious and non-infectious models revealed that B lymphocytes could regulate and play crucial roles in cellular responses. Here, we analyze the trypomastigote-induced effect on B cells, their effects on CD4+ T cells, and their correlation with in vivo findings. The trypomastigotes were able to induce the proliferation and the production of IL-10 or IL-6 of naïve B cells in co-culture experiments. Also, we found that IL-10-producing B220lo cells were elicited in vivo. We also found up-regulated expression of FasL and PD-L1, proteins involved in apoptosis induction and inhibition of TCR signaling, and of BAFF and APRIL mRNAs, two B-cell growth factors. Interestingly, it was observed that IL-21, which plays a critical role in regulatory B cell differentiation, was significantly increased in B220+/IL-21+ in in vivo infections. This is striking since the secretion of IL-21 is associated with T helper follicular cells. Furthermore, trypomastigote-stimulated B-cell conditioned medium dramatically reduced the proliferation and increased the apoptotic rate on CD3/CD28 activated CD4+ T cells, suggesting the development of effective regulatory B cells. In this condition, CD4+ T cells showed a marked decrease in proliferation and viability with marginal IL-2 or IFNγ secretion, which is counterproductive with an efficient immune response against T. cruzi. Altogether, our results show that B lymphocytes stimulated with trypomastigotes adopt a particular phenotype that exerts a strong regulation of this T cell compartment by inducing apoptosis, arresting cell division, and affecting the developing of a proinflammatory response.
Collapse
Affiliation(s)
- Martín Somoza
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín-CONICET, Buenos Aires, Argentina
| | - Adriano Bertelli
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín-CONICET, Buenos Aires, Argentina
| | - Cecilia A. Pratto
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín-CONICET, Buenos Aires, Argentina
| | - Ramiro E. Verdun
- Sylvester Comprehensive Cancer Center and Division of Hematology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Oscar Campetella
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín-CONICET, Buenos Aires, Argentina
| | - Juan Mucci
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín-CONICET, Buenos Aires, Argentina
| |
Collapse
|
5
|
Goodall CP, Schwarz B, Selivanovitch E, Avera J, Wang J, Miettinen H, Douglas T. Controlled Modular Multivalent Presentation of the CD40 Ligand on P22 Virus-like Particles Leads to Tunable Amplification of CD40 Signaling. ACS APPLIED BIO MATERIALS 2021; 4:8205-8214. [PMID: 35005938 DOI: 10.1021/acsabm.1c00718] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ligands of the tumor necrosis factor superfamily (TNFSF) are appealing targets for immunotherapy research due to their integral involvement in stimulation or restriction of immune responses. TNFSF-targeted therapies are currently being developed to combat immunologically based diseases and cancer. A crucial determinant of effective TNFSF receptor binding and signaling is the trimeric quaternary structure of the ligand. Additionally, ligand multivalency is essential to propagate strong signaling in effector cells. Thus, designing a synthetic platform to display trimeric TNFSF ligands in a multivalent manner is necessary to further the understanding of ligand-receptor interactions. Viral nanocages have architectures that are amenable to genetic and chemical modifications of both their interior and exterior surfaces. Notably, the exterior surface of virus-like particles can be utilized as a platform for the modular multivalent presentation of target proteins. In this study, we build on previous efforts exploring the bacteriophage P22 virus-like particle for the exterior multivalent modular display of a potent immune-stimulating TNFSF protein, CD40 ligand (CD40L). Using a cell-based reporter system, we quantify the effects of tunable avidity on CD40 signaling by CD40L displayed on the surface of P22 nanocages. Multivalent presentation of CD40L resulted in a 53.6-fold decrease of the half maximal effective concentration (EC50) compared to free CD40L, indicating higher potency. Our results emphasize the power of using P22-based biomimetics to study ligand-receptor interactions within their proper structural context, which may contribute to the development of effective immune modulators.
Collapse
Affiliation(s)
- Cheri Peyton Goodall
- Department of Chemistry, Indiana University, 800 East Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - Benjamin Schwarz
- Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, 903 South 4th Street, Hamilton, Montana 59840, United States
| | - Ekaterina Selivanovitch
- Department of Chemistry, Indiana University, 800 East Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - John Avera
- Walden Biosciences, One Kendall Square, Suite 7102, Cambridge, Massachusetts 02139, United States
| | - Joseph Wang
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, 700 HMC Crescent Road, Hershey, Pennsylvania 17033, United States
| | - Heini Miettinen
- Department of Microbiology and Immunology, Montana State University, P.O. Box 173520, Bozeman, Montana 59717, United States
| | - Trevor Douglas
- Department of Chemistry, Indiana University, 800 East Kirkwood Avenue, Bloomington, Indiana 47405, United States
| |
Collapse
|
6
|
Koch PD, Pittet MJ, Weissleder R. The chemical biology of IL-12 production via the non-canonical NFkB pathway. RSC Chem Biol 2020; 1:166-176. [PMID: 34458756 PMCID: PMC8341911 DOI: 10.1039/d0cb00022a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022] Open
Abstract
Interleukin-12 (IL-12) has emerged as an attractive cytokine for cancer therapy because it has direct anti-cancer effects and additionally plays a critical role in enhancing checkpoint inhibitors. Given these multiple modes of actions, identifying means to pharmacologically induce IL-12 production in the tumor microenvironment has become important. In this review, we highlight therapeutics that promote IL-12 induction in tumor-associated myeloid cells through the non-canonical NFkB pathway. We discuss existing clinical trials and briefly examine the additional pathway targets that warrant further exploration for drug discovery.
Collapse
Affiliation(s)
- Peter D Koch
- Center for Systems Biology, Massachusetts General Hospital 185 Cambridge St Boston MA 02114 USA
- Department of Systems Biology, Harvard Medical School 200 Longwood Ave Boston MA 02115 USA
| | - Mikael J Pittet
- Center for Systems Biology, Massachusetts General Hospital 185 Cambridge St Boston MA 02114 USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital 185 Cambridge St Boston MA 02114 USA
- Department of Systems Biology, Harvard Medical School 200 Longwood Ave Boston MA 02115 USA
| |
Collapse
|
7
|
Lee Y, Chung B, Ko D, Lim HS. A solid-phase method for synthesis of dimeric and trimeric ligands: Identification of potent bivalent ligands of 14-3-3σ. Bioorg Chem 2019; 91:103141. [DOI: 10.1016/j.bioorg.2019.103141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/12/2019] [Accepted: 07/19/2019] [Indexed: 01/12/2023]
|
8
|
Wennhold K, Shimabukuro-Vornhagen A, von Bergwelt-Baildon M. B Cell-Based Cancer Immunotherapy. Transfus Med Hemother 2019; 46:36-46. [PMID: 31244580 PMCID: PMC6558332 DOI: 10.1159/000496166] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 12/10/2018] [Indexed: 12/13/2022] Open
Abstract
B cells are not only producers of antibodies, but also contribute to immune regulation or act as potent antigen-presenting cells. The potential of B cells for cellular therapy is still largely underestimated, despite their multiple diverse effector functions. The CD40L/CD40 signaling pathway is the most potent activator of antigen presentation capacity in B lymphocytes. CD40-activated B cells are potent antigen-presenting cells that induce specific T-cell responses in vitro and in vivo. In preclinical cancer models in mice and dogs, CD40-activated B cell-based cancer immunotherapy was able to induce effective antitumor immunity. So far, there have been only few early-stage clinical studies involving B cell-based cancer vaccines. These trials indicate that B cell-based immunotherapy is generally safe and associated with little toxicity. Furthermore, these studies suggest that B-cell immunotherapy can elicit antitumor T-cell responses. Alongside the recent advances in cellular therapies in general, major obstacles for generation of good manufacturing practice-manufactured B-cell immunotherapies have been overcome. Thus, a first clinical trial involving CD40-activated B cells might be in reach.
Collapse
Affiliation(s)
- Kerstin Wennhold
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | | | - Michael von Bergwelt-Baildon
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Gene Center Munich, LMU Munich, Munich, Germany
| |
Collapse
|
9
|
Yang S, Wang J, Brand DD, Zheng SG. Role of TNF-TNF Receptor 2 Signal in Regulatory T Cells and Its Therapeutic Implications. Front Immunol 2018; 9:784. [PMID: 29725328 PMCID: PMC5916970 DOI: 10.3389/fimmu.2018.00784] [Citation(s) in RCA: 240] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 03/28/2018] [Indexed: 12/24/2022] Open
Abstract
Tumor necrosis factor α (TNFα) is a pleiotropic cytokine which signals through TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2). Emerging evidence has demonstrated that TNFR1 is ubiquitously expressed on almost all cells, while TNFR2 exhibits a limited expression, predominantly on regulatory T cells (Tregs). In addition, the signaling pathway by sTNF via TNFR1 mainly triggers pro-inflammatory pathways, and mTNF binding to TNFR2 usually initiates immune modulation and tissue regeneration. TNFα plays a critical role in upregulation or downregulation of Treg activity. Deficiency in TNFR2 signaling is significant in various autoimmune diseases. An ideal therapeutic strategy for autoimmune diseases would be to selectively block the sTNF/TNFR1 signal through the administration of sTNF inhibitors, or using TNFR1 antagonists while keeping the TNFR2 signaling pathway intact. Another promising strategy would be to rely on TNFR2 agonists which could drive the expansion of Tregs and promote tissue regeneration. Design of these therapeutic strategies targeting the TNFR1 or TNFR2 signaling pathways holds promise for the treatment of diverse inflammatory and degenerative diseases.
Collapse
Affiliation(s)
- Sujuan Yang
- Department of Clinical Immunology, Third Hospital at Sun Yat-sen University, Guangzhou, China.,Division of Rheumatology, Milton S. Hershey Medical Center at Penn State University, Hershey, PA, United States
| | - Julie Wang
- Division of Rheumatology, Milton S. Hershey Medical Center at Penn State University, Hershey, PA, United States
| | | | - Song Guo Zheng
- Division of Rheumatology, Milton S. Hershey Medical Center at Penn State University, Hershey, PA, United States
| |
Collapse
|
10
|
Kozel V, Daniliuc CG, Kirsch P, Haufe G. C 3 -Symmetric Tricyclo[2.2.1.0 2,6 ]heptane-3,5,7-triol. Angew Chem Int Ed Engl 2017; 56:15456-15460. [PMID: 29044954 DOI: 10.1002/anie.201709279] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Indexed: 11/06/2022]
Abstract
A straightforward access to a hitherto unknown C3 -symmetric tricyclic triol both in racemic and enantiopure forms has been developed. Treatment of 7-tert-butoxynorbornadiene with peroxycarboxylic acids provided mixtures of C1 - and C3 -symmetric 3,5,7-triacyloxynortricyclenes via transannular π-cyclization and replacement of the tert-butoxy group. By refluxing in formic acid, the C1 -symmetric esters were converted to the C3 -symmetric formate. Hydrolysis gave diastereoisomeric triols, which were separated by recrystallization. Enantiomer resolution via diastereoisomeric tri(O-methylmandelates) delivered the target triols on a gram scale. The pure enantiomers are useful as core units of dopants for liquid crystals.
Collapse
Affiliation(s)
- Volodymyr Kozel
- Organisch-Chemisches Institut, Universität Münster, Corrensstrasse 40, 48149, Münster, Germany
| | | | - Peer Kirsch
- Integrated Circuit Materials R&D Germany, Merck KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Günter Haufe
- Organisch-Chemisches Institut, Universität Münster, Corrensstrasse 40, 48149, Münster, Germany
| |
Collapse
|
11
|
Chen J, Song Y, Bojadzic D, Tamayo-Garcia A, Landin AM, Blomberg BB, Buchwald P. Small-Molecule Inhibitors of the CD40-CD40L Costimulatory Protein-Protein Interaction. J Med Chem 2017; 60:8906-8922. [PMID: 29024591 PMCID: PMC5823691 DOI: 10.1021/acs.jmedchem.7b01154] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Costimulatory interactions are required for T cell activation and development of an effective immune response; hence, they are valuable therapeutic targets for immunomodulation. However, they, as all other protein-protein interactions, are difficult to target by small molecules. Here, we report the identification of novel small-molecule inhibitors of the CD40-CD40L interaction designed starting from the chemical space of organic dyes. For the most promising compounds such as DRI-C21045, activity (IC50) in the low micromolar range has been confirmed in cell assays including inhibition of CD40L-induced activation in NF-κB sensor cells, THP-1 myeloid cells, and primary human B cells as well as in murine allogeneic skin transplant and alloantigen-induced T cell expansion in draining lymph node experiments. Specificity versus other TNF-superfamily interactions (TNF-R1-TNF-α) and lack of cytotoxicity have also been confirmed at these concentrations. These novel compounds provide proof-of-principle evidence for the possibility of small-molecule inhibition of costimulatory protein-protein interactions, establish the structural requirements needed for efficient CD40-CD40L inhibition, and serve to guide the search for such immune therapeutics.
Collapse
Affiliation(s)
- Jinshui Chen
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| | - Yun Song
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
- Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| | - Damir Bojadzic
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| | - Alejandro Tamayo-Garcia
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| | - Ana Marie Landin
- Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| | - Bonnie B. Blomberg
- Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
- Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| |
Collapse
|
12
|
Kozel V, Daniliuc CG, Kirsch P, Haufe G. Das C
3
-symmetrische Tricyclo[2.2.1.02,6
]heptan-3,5,7-triol. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201709279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Volodymyr Kozel
- Organisch-Chemisches Institut; Universität Münster; Corrensstraße 40 48149 Münster Deutschland
| | | | - Peer Kirsch
- Integrated Circuit Materials R&D Germany; Merck KGaA; Frankfurter Straße 250 64293 Darmstadt Deutschland
| | - Günter Haufe
- Organisch-Chemisches Institut; Universität Münster; Corrensstraße 40 48149 Münster Deutschland
| |
Collapse
|
13
|
Martin C, Waghela SD, Lokhandwala S, Ambrus A, Bray J, Vuong C, Vinodkumar V, Dominowski PJ, Rai S, Mwangi D, Foss DL, Mwangi W. Characterization of a Broadly Reactive Anti-CD40 Agonistic Monoclonal Antibody for Potential Use as an Adjuvant. PLoS One 2017; 12:e0170504. [PMID: 28107431 PMCID: PMC5249191 DOI: 10.1371/journal.pone.0170504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 01/05/2017] [Indexed: 01/22/2023] Open
Abstract
Lack of safe and effective adjuvants is a major hindrance to the development of efficacious vaccines. Signaling via CD40 pathway leads to enhanced antigen processing and presentation, nitric oxide expression, pro-inflammatory cytokine expression by antigen presenting cells, and stimulation of B-cells to undergo somatic hypermutation, immunoglobulin class switching, and proliferation. Agonistic anti-CD40 antibodies have shown promising adjuvant qualities in human and mouse vaccine studies. An anti-CD40 monoclonal antibody (mAb), designated 2E4E4, was identified and shown to have strong agonistic effects on primary cells from multiple livestock species. The mAb recognize swine, bovine, caprine, and ovine CD40, and evoked 25-fold or greater proliferation of peripheral blood mononuclear cells (PBMCs) from these species relative to cells incubated with an isotype control (p<0.001). In addition, the mAb induced significant nitric oxide (p<0.0001) release by bovine macrophages. Furthermore, the mAb upregulated the expression of MHC-II by PBMCs, and stimulated significant (p<0.0001) IL-1α, IL6, IL-8, and TNF-α expression by PBMCs. These results suggest that the mAb 2E4E4 can target and stimulate cells from multiple livestock species and thus, it is a potential candidate for adjuvant development. This is the first study to report an anti-swine CD40 agonistic mAb that is also broadly reactive against multiple species.
Collapse
Affiliation(s)
- Cameron Martin
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Suryakant D. Waghela
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Shehnaz Lokhandwala
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Andy Ambrus
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Jocelyn Bray
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Christina Vuong
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Vanitha Vinodkumar
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | | | - Sharath Rai
- Zoetis, Kalamazoo, Michigan, United States of America
| | - Duncan Mwangi
- Zoetis, Kalamazoo, Michigan, United States of America
| | | | - Waithaka Mwangi
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
14
|
Mándity IM, Fülöp F. An overview of peptide and peptoid foldamers in medicinal chemistry. Expert Opin Drug Discov 2015; 10:1163-77. [PMID: 26289578 DOI: 10.1517/17460441.2015.1076790] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Foldamers are artificial self-organizing systems with various critical properties: i) a stable and designable secondary structure; ii) a larger molecular surface as compared with ordinary organic drug molecules; iii) appropriate control of the orientation of the side-chain functional groups; iv) resistance against proteolytic degradation, which leads to potentially increased oral bioavailability and a longer serum half-life relative to ordinary α-peptides; and v) the lower conformational freedom may result in increased receptor binding in comparison with the natural analogs. AREAS COVERED This article covers the general properties and types of foldamers. This includes highlighted examples of medicinal chemical applications, including antibacterial and cargo molecules, anti-Alzheimer compounds and protein-protein interaction modifiers. EXPERT OPINION Various new foldamers have been created with a range of structures and biological applications. Membrane-acting antibacterial foldamers have been introduced. A general property of these structures is their amphiphilic nature. The amphiphilicity can be stationary or induced by the membrane binding. Cell-penetrating foldamers have been described which serve as cargo molecules, and foldamers have been used as autophagy inducers. Anti-Alzheimer compounds too have been created and the greatest breakthrough was attained via the modification of protein-protein interactions. This can serve as the chemical and pharmaceutical basis for the relevance of foldamers in the future.
Collapse
Affiliation(s)
| | - Ferenc Fülöp
- a University of Szeged Institute of Pharmaceutical Chemistry , H-6720 Szeged, Eötvös u. 6, Hungary +36 62 545 768 ; +36 62 545 564 ; +36 62 545 705 ; ;
| |
Collapse
|
15
|
Russier J, Grillaud M, Bianco A. Elucidation of the Cellular Uptake Mechanisms of Polycationic HYDRAmers. Bioconjug Chem 2015; 26:1484-93. [DOI: 10.1021/acs.bioconjchem.5b00270] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Julie Russier
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunopathologie et Chimie Thérapeutique, 67000 Strasbourg, France
| | - Maxime Grillaud
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunopathologie et Chimie Thérapeutique, 67000 Strasbourg, France
| | - Alberto Bianco
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunopathologie et Chimie Thérapeutique, 67000 Strasbourg, France
| |
Collapse
|
16
|
Ding Q, Si X, Liu D, Peng J, Tang H, Sun W, Rui M, Chen Q, Wu L, Xu Y. Targeting and liposomal drug delivery to CD40L expressing T cells for treatment of autoimmune diseases. J Control Release 2015; 207:86-92. [PMID: 25839125 DOI: 10.1016/j.jconrel.2015.03.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 03/05/2015] [Accepted: 03/30/2015] [Indexed: 10/23/2022]
Abstract
CD40L is considered as an important target for the treatment of autoimmune diseases. There have been many efforts devoted to the development of antibodies and other molecules to disrupt CD40/CD40L interaction for therapeutic benefits. In this study, we designed a CD40L specific peptide ligand - A25 based on CD40L crystal structure and molecular docking studies. Its binding affinity and specificity to CD40L were confirmed by Surface Plasmon Resonance (SPR) measurements. The peptide A25 was then conjugated on the surface of liposomes and shown to be able to mediate specific liposomal drug delivery to CD40L+ cells. Loaded with the cytostatic drug methotrexate (MTX), the A25 modified liposome could significantly reduce the CD40L+ cell ratios in the experimental autoimmune encephalomyelitis (EAE) mice, resulting in great improvement in clinical scores. Since CD40L+ cells are involved in the pathological development of many auto-immune diseases, A25 conjugated drug targeting systems may be useful for developing therapies that are more efficacies and with less side effects.
Collapse
Affiliation(s)
- Qian Ding
- School of Pharmacy, Med-X Research Institute and School of Biomedical, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, PR China
| | - Xiaofei Si
- School of Pharmacy, Med-X Research Institute and School of Biomedical, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, PR China
| | - Dan Liu
- School of Pharmacy, Med-X Research Institute and School of Biomedical, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, PR China
| | - Jinliang Peng
- School of Pharmacy, Med-X Research Institute and School of Biomedical, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, PR China
| | - Hailing Tang
- School of Pharmacy, Med-X Research Institute and School of Biomedical, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, PR China
| | - Wenqiang Sun
- School of Pharmacy, Med-X Research Institute and School of Biomedical, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, PR China
| | - Mengjie Rui
- School of Pharmacy, Med-X Research Institute and School of Biomedical, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, PR China
| | - Qunli Chen
- School of Pharmacy, Med-X Research Institute and School of Biomedical, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, PR China
| | - Lieyi Wu
- School of Pharmacy, Med-X Research Institute and School of Biomedical, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, PR China
| | - Yuhong Xu
- School of Pharmacy, Med-X Research Institute and School of Biomedical, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, PR China.
| |
Collapse
|
17
|
Grillaud M, Bianco A. Multifunctional adamantane derivatives as new scaffolds for the multipresentation of bioactive peptides. J Pept Sci 2014; 21:330-45. [DOI: 10.1002/psc.2719] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/16/2014] [Accepted: 10/20/2014] [Indexed: 12/31/2022]
Affiliation(s)
- Maxime Grillaud
- CNRS, Institut de Biologie Moléculaire et; Cellulaire, Immunopathologie et Chimie Thérapeutique; Strasbourg France
| | - Alberto Bianco
- CNRS, Institut de Biologie Moléculaire et; Cellulaire, Immunopathologie et Chimie Thérapeutique; Strasbourg France
| |
Collapse
|
18
|
Lamanna G, Grillaud M, Macri C, Chaloin O, Muller S, Bianco A. Adamantane-based dendrons for trimerization of the therapeutic P140 peptide. Biomaterials 2014; 35:7553-61. [DOI: 10.1016/j.biomaterials.2014.05.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/09/2014] [Indexed: 12/21/2022]
|
19
|
Wennhold K, Shimabukuro-Vornhagen A, Theurich S, von Bergwelt-Baildon M. CD40-activated B cells as antigen-presenting cells: the final sprint toward clinical application. Expert Rev Vaccines 2013; 12:631-7. [PMID: 23750793 DOI: 10.1586/erv.13.39] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Efficient antigen presentation is a prerequisite for the development of a T-cell-mediated immune response in vitro and in vivo. CD40-activated B cells (CD40B cells) are a promising alternative to dendritic cells as professional APCs for immunotherapy. CD40 activation dramatically improves antigen presentation by normal and malignant B cells, efficiently inducing naive and memory CD4(+) and CD8(+) T-cell responses. Moreover, CD40B cells do not only attract T cells by release of chemokines, but also home to secondary lymphoid organs. Furthermore, CD40B cells can be expanded exponentially over several weeks at high purity without a loss of antigen-presenting function, providing an almost unlimited source of cellular adjuvant. Vaccination with CD40B cells was shown in mice and dogs to induce a specific immune response. This article summarizes the achievements of intense research on CD40B cells over the last decade, as well as novel developments critical for a rapid translation into clinical application.
Collapse
Affiliation(s)
- Kerstin Wennhold
- Cologne Interventional Immunology, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany.
| | | | | | | |
Collapse
|
20
|
Ulrich S, Dumy P, Boturyn D, Renaudet O. Engineering of biomolecules for sensing and imaging applications. J Drug Deliv Sci Technol 2013. [DOI: 10.1016/s1773-2247(13)50001-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
21
|
Levine PM, Carberry TP, Holub JM, Kirshenbaum K. Crafting precise multivalent architectures. MEDCHEMCOMM 2013. [DOI: 10.1039/c2md20338c] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Development of elaborate three-dimensional multivalent displays appended on natural or synthetic molecular scaffolds.
Collapse
|
22
|
Levine PM, Lee E, Greenfield A, Bonneau R, Logan SK, Garabedian MJ, Kirshenbaum K. Androgen receptor antagonism by divalent ethisterone conjugates in castrate-resistant prostate cancer cells. ACS Chem Biol 2012; 7:1693-701. [PMID: 22871957 DOI: 10.1021/cb300332w] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Sustained treatment of prostate cancer with androgen receptor (AR) antagonists can evoke drug resistance, leading to castrate-resistant disease. Elevated activity of the AR is often associated with this highly aggressive disease state. Therefore, new therapeutic regimens that target and modulate AR activity could prove beneficial. We previously introduced a versatile chemical platform to generate competitive and non-competitive multivalent peptoid oligomer conjugates that modulate AR activity. In particular, we identified a linear and a cyclic divalent ethisterone conjugate that exhibit potent anti-proliferative properties in LNCaP-abl cells, a model of castrate-resistant prostate cancer. Here, we characterize the mechanism of action of these compounds utilizing confocal microscopy, time-resolved fluorescence resonance energy transfer, chromatin immunoprecipitation, flow cytometry, and microarray analysis. The linear conjugate competitively blocks AR action by inhibiting DNA binding. In addition, the linear conjugate does not promote AR nuclear localization or co-activator binding. In contrast, the cyclic conjugate promotes AR nuclear localization and induces cell-cycle arrest, despite its inability to compete against endogenous ligand for binding to AR in vitro. Genome-wide expression analysis reveals that gene transcripts are differentially affected by treatment with the linear or cyclic conjugate. Although the divalent ethisterone conjugates share extensive chemical similarities, we illustrate that they can antagonize the AR via distinct mechanisms of action, establishing new therapeutic strategies for potential applications in AR pharmacology.
Collapse
Affiliation(s)
- Paul M. Levine
- Department
of Chemistry and §Center for Genomics and Systems Biology, New York University, New York, New York 10003, United
States
- Department
of Biochemistry and Molecular Pharmacology, ⊥Department of Urology, and ∥Department of
Microbiology, NYU Langone School of Medicine, New York, New York 10016, United States
| | - Eugine Lee
- Department
of Chemistry and §Center for Genomics and Systems Biology, New York University, New York, New York 10003, United
States
- Department
of Biochemistry and Molecular Pharmacology, ⊥Department of Urology, and ∥Department of
Microbiology, NYU Langone School of Medicine, New York, New York 10016, United States
| | - Alex Greenfield
- Department
of Chemistry and §Center for Genomics and Systems Biology, New York University, New York, New York 10003, United
States
- Department
of Biochemistry and Molecular Pharmacology, ⊥Department of Urology, and ∥Department of
Microbiology, NYU Langone School of Medicine, New York, New York 10016, United States
| | - Richard Bonneau
- Department
of Chemistry and §Center for Genomics and Systems Biology, New York University, New York, New York 10003, United
States
- Department
of Biochemistry and Molecular Pharmacology, ⊥Department of Urology, and ∥Department of
Microbiology, NYU Langone School of Medicine, New York, New York 10016, United States
| | - Susan K. Logan
- Department
of Chemistry and §Center for Genomics and Systems Biology, New York University, New York, New York 10003, United
States
- Department
of Biochemistry and Molecular Pharmacology, ⊥Department of Urology, and ∥Department of
Microbiology, NYU Langone School of Medicine, New York, New York 10016, United States
| | - Michael J. Garabedian
- Department
of Chemistry and §Center for Genomics and Systems Biology, New York University, New York, New York 10003, United
States
- Department
of Biochemistry and Molecular Pharmacology, ⊥Department of Urology, and ∥Department of
Microbiology, NYU Langone School of Medicine, New York, New York 10016, United States
| | - Kent Kirshenbaum
- Department
of Chemistry and §Center for Genomics and Systems Biology, New York University, New York, New York 10003, United
States
- Department
of Biochemistry and Molecular Pharmacology, ⊥Department of Urology, and ∥Department of
Microbiology, NYU Langone School of Medicine, New York, New York 10016, United States
| |
Collapse
|
23
|
Stein F, Mehmood T, Plass T, Zaidi JH, Diederichsen U. Synthesis of trifunctional cyclo-β-tripeptide templates. Beilstein J Org Chem 2012; 8:1576-83. [PMID: 23209489 PMCID: PMC3510989 DOI: 10.3762/bjoc.8.180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 08/20/2012] [Indexed: 12/17/2022] Open
Abstract
The concept of template-assembled synthetic proteins (TASP) describes a central scaffold that predefines the three dimensional structure for diverse molecules linked to this platform. Cyclic β-tripeptides are interesting candidates for use as templates due to their conformationally defined structure, stability to enzymatic degradation, and ability to form intermolecular stacked tubular structures. To validate the applicability of cyclic β-tripeptides within the TASP concept, an efficient synthesis of the cyclopeptide with orthogonal functionalization of the side chains is desired. A solid-phase-supported route with on-resin cyclization is described, employing the aryl hydrazide linker cleavable by oxidation. An orthogonal protection-group strategy allows functionalization of the central cyclic β-tripeptide with up to three different peptide fragments or fluorescent labels.
Collapse
Affiliation(s)
- Frank Stein
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Tammannstrasse 2, D-37077 Göttingen, Germany
| | - Tahir Mehmood
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Tammannstrasse 2, D-37077 Göttingen, Germany
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Tilman Plass
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Tammannstrasse 2, D-37077 Göttingen, Germany
| | - Javid H Zaidi
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Ulf Diederichsen
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Tammannstrasse 2, D-37077 Göttingen, Germany
| |
Collapse
|
24
|
Fülöp L, Mándity IM, Juhász G, Szegedi V, Hetényi A, Wéber E, Bozsó Z, Simon D, Benkő M, Király Z, Martinek TA. A foldamer-dendrimer conjugate neutralizes synaptotoxic β-amyloid oligomers. PLoS One 2012; 7:e39485. [PMID: 22859942 PMCID: PMC3408453 DOI: 10.1371/journal.pone.0039485] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 05/21/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND AIMS Unnatural self-organizing biomimetic polymers (foldamers) emerged as promising materials for biomolecule recognition and inhibition. Our goal was to construct multivalent foldamer-dendrimer conjugates which wrap the synaptotoxic β-amyloid (Aβ) oligomers with high affinity through their helical foldamer tentacles. Oligomeric Aβ species play pivotal role in Alzheimer's disease, therefore recognition and direct inhibition of this undruggable target is a great current challenge. METHODS AND RESULTS Short helical β-peptide foldamers with designed secondary structures and side chain chemistry patterns were applied as potential recognition segments and their binding to the target was tested with NMR methods (saturation transfer difference and transferred-nuclear Overhauser effect). Helices exhibiting binding in the µM region were coupled to a tetravalent G0-PAMAM dendrimer. In vitro biophysical (isothermal titration calorimetry, dynamic light scattering, transmission electron microscopy and size-exclusion chromatography) and biochemical tests (ELISA and dot blot) indicated the tight binding between the foldamer conjugates and the Aβ oligomers. Moreover, a selective low nM interaction with the low molecular weight fraction of the Aβ oligomers was found. Ex vivo electrophysiological experiments revealed that the new material rescues the long-term potentiation from the toxic Aβ oligomers in mouse hippocampal slices at submicromolar concentration. CONCLUSIONS The combination of the foldamer methodology, the fragment-based approach and the multivalent design offers a pathway to unnatural protein mimetics that are capable of specific molecular recognition, and has already resulted in an inhibitor for an extremely difficult target.
Collapse
Affiliation(s)
- Lívia Fülöp
- Department of Medical Chemistry, University of Szeged, Szeged, Hungary
| | - István M. Mándity
- Institute of Pharmaceutical Chemistry, University of Szeged, Szeged, Hungary
| | - Gábor Juhász
- Department of Medical Chemistry, University of Szeged, Szeged, Hungary
| | - Viktor Szegedi
- Bay Zoltán Foundation for Applied Research – BAYGEN, Szeged, Hungary
| | | | - Edit Wéber
- Institute of Pharmaceutical Chemistry, University of Szeged, Szeged, Hungary
| | - Zsolt Bozsó
- Department of Medical Chemistry, University of Szeged, Szeged, Hungary
| | - Dóra Simon
- Department of Medical Chemistry, University of Szeged, Szeged, Hungary
| | - Mária Benkő
- Department of Physical Chemistry and Materials Science, University of Szeged, Szeged, Hungary
| | - Zoltán Király
- Department of Physical Chemistry and Materials Science, University of Szeged, Szeged, Hungary
| | - Tamás A. Martinek
- Institute of Pharmaceutical Chemistry, University of Szeged, Szeged, Hungary
| |
Collapse
|
25
|
Yoshinari T, Gessier F, Noti C, Beck AK, Seebach D. Stereoselective Preparation of 3-Amino-2-fluoro Carboxylic Acid Derivatives, and Their Incorporation in Tetrahydropyrimidin-4(1H)-ones, and in Open-Chain and Cyclic β-Peptides. Helv Chim Acta 2011. [DOI: 10.1002/hlca.201100340] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
26
|
Silvian LF, Friedman JE, Strauch K, Cachero TG, Day ES, Qian F, Cunningham B, Fung A, Sun L, Shipps GW, Su L, Zheng Z, Kumaravel G, Whitty A. Small molecule inhibition of the TNF family cytokine CD40 ligand through a subunit fracture mechanism. ACS Chem Biol 2011; 6:636-47. [PMID: 21417339 PMCID: PMC3415792 DOI: 10.1021/cb2000346] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BIO8898 is one of several synthetic organic molecules that have recently been reported to inhibit receptor binding and function of the constitutively trimeric tumor necrosis factor (TNF) family cytokine CD40 ligand (CD40L, aka CD154). Small molecule inhibitors of protein-protein interfaces are relatively rare, and their discovery is often very challenging. Therefore, to understand how BIO8898 achieves this feat, we characterized its mechanism of action using biochemical assays and X-ray crystallography. BIO8898 inhibited soluble CD40L binding to CD40-Ig with a potency of IC(50) = 25 μM and inhibited CD40L-dependent apoptosis in a cellular assay. A co-crystal structure of BIO8898 with CD40L revealed that one inhibitor molecule binds per protein trimer. Surprisingly, the compound binds not at the surface of the protein but by intercalating deeply between two subunits of the homotrimeric cytokine, disrupting a constitutive protein-protein interface and breaking the protein's 3-fold symmetry. The compound forms several hydrogen bonds with the protein, within an otherwise hydrophobic binding pocket. In addition to the translational splitting of the trimer, binding of BIO8898 was accompanied by additional local and longer-range conformational perturbations of the protein, both in the core and in a surface loop. Binding of BIO8898 is reversible, and the resulting complex is stable and does not lead to detectable dissociation of the protein trimer. Our results suggest that a set of core aromatic residues that are conserved across a subset of TNF family cytokines might represent a generic hot-spot for the induced-fit binding of trimer-disrupting small molecules.
Collapse
Affiliation(s)
- Laura F. Silvian
- Department of Drug Discovery, Biogen Idec, 12 Cambridge Center, Cambridge, Massachusetts 02142.
,To whom correspondence should be addressed: ,
| | - Jessica E. Friedman
- Department of Drug Discovery, Biogen Idec, 12 Cambridge Center, Cambridge, Massachusetts 02142.
| | - Kathy Strauch
- Department of Drug Discovery, Biogen Idec, 12 Cambridge Center, Cambridge, Massachusetts 02142.
| | - Teresa G. Cachero
- Department of Drug Discovery, Biogen Idec, 12 Cambridge Center, Cambridge, Massachusetts 02142.
| | - Eric S. Day
- Department of Drug Discovery, Biogen Idec, 12 Cambridge Center, Cambridge, Massachusetts 02142.
| | - Fang Qian
- Department of Drug Discovery, Biogen Idec, 12 Cambridge Center, Cambridge, Massachusetts 02142.
| | - Brian Cunningham
- Sunesis Pharmaceuticals, Incorporated, 341 Oyster Point Boulevard, South San Francisco, CA 94080.
| | - Amy Fung
- Sunesis Pharmaceuticals, Incorporated, 341 Oyster Point Boulevard, South San Francisco, CA 94080.
| | - Lihong Sun
- Department of Drug Discovery, Biogen Idec, 12 Cambridge Center, Cambridge, Massachusetts 02142.
| | - Gerald W. Shipps
- Neogenesis Pharmaceuticals Inc., 840 Memorial Dr., Cambridge, MA 02139
| | - Lihe Su
- Department of Drug Discovery, Biogen Idec, 12 Cambridge Center, Cambridge, Massachusetts 02142.
| | - Zhongli Zheng
- Department of Drug Discovery, Biogen Idec, 12 Cambridge Center, Cambridge, Massachusetts 02142.
| | | | - Adrian Whitty
- Boston University, Department of Chemistry, Metcalf Center for Science and Engineering, 590 Commonwealth Ave, Boston, MA 02215.
,To whom correspondence should be addressed: ,
| |
Collapse
|
27
|
Shin SBY, Almeida RD, Gerona-Navarro G, Bracken C, Jaffrey SR. Assembling ligands in situ using bioorthogonal boronate ester synthesis. ACTA ACUST UNITED AC 2011; 17:1171-6. [PMID: 21095566 DOI: 10.1016/j.chembiol.2010.09.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 08/19/2010] [Accepted: 09/09/2010] [Indexed: 10/18/2022]
Abstract
Many molecules that could manipulate cellular function are not practical due to their large size and concomitant undesirable pharmocokinetic properties. Here, we describe a bioorthogonal, highly stable boronate ester (HiSBE) synthesis and use this reaction to synthesize a biologically active molecule from smaller precursors in a physiological context. The rapid rate of HiSBE synthesis suggests that it may be useful for assembling a wide variety of biologically active molecules in physiological solutions.
Collapse
Affiliation(s)
- Sung Bin Y Shin
- Department of Pharmacology, Weill Medical College, Cornell University, New York, NY 10065, USA
| | | | | | | | | |
Collapse
|
28
|
Faustman D, Davis M. TNF receptor 2 pathway: drug target for autoimmune diseases. Nat Rev Drug Discov 2010; 9:482-93. [PMID: 20489699 DOI: 10.1038/nrd3030] [Citation(s) in RCA: 317] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although drug development has advanced for autoimmune diseases, many current therapies are hampered by adverse effects and the frequent destruction or inactivation of healthy cells in addition to pathological cells. Targeted autoimmune therapies capable of eradicating the rare autoreactive immune cells that are responsible for the attack on the body's own cells are yet to be identified. This Review presents a new emerging approach aimed at selectively destroying autoreactive immune cells by specific activation of tumour necrosis factor receptor 2 (TNFR2), which is found on autoreactive and normal T lymphocytes, with the potential of avoiding or reducing the toxicity observed with existing therapies.
Collapse
Affiliation(s)
- Denise Faustman
- Immunobiology Laboratory, Room 3602, Building 149, Massachusetts General Hospital and Harvard Medical School, 13th Street, Charlestown, Massachusetts 02129, USA.
| | | |
Collapse
|
29
|
Buchwald P, Margolles-Clark E, Kenyon NS, Ricordi C. Organic dyes as small molecule protein-protein interaction inhibitors for the CD40-CD154 costimulatory interaction. J Mol Recognit 2010; 23:65-73. [PMID: 19621420 DOI: 10.1002/jmr.969] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is becoming increasingly clear that small molecules can often act as effective protein-protein interaction (PPI) inhibitors, an area of increasing interest for its many possible therapeutic applications. We have identified several organic dyes and related small molecules that (i) concentration-dependently inhibit the important CD40-CD154 costimulatory interaction with activities in the low micromolar (microM) range, (ii) show selectivity toward this particular PPI, (iii) seem to bind on the surface of CD154, and (iv) concentration-dependently inhibit the CD154-induced B cell proliferation. They were identified through an iterative activity screening/structural similarity search procedure starting with suramin as lead, and the best smaller compounds, the main focus of the present work, achieved an almost 3-fold increase in ligand efficiency (DeltaG(0)/nonhydrogen atom = 0.8 kJ/N(nHa)) approaching the average of known promising small-molecule PPI inhibitors (approximately 1.0 kJ/N(nHa)). Since CD154 is a member of the tumor necrosis factor (TNF) superfamily of cell surface interaction molecules, inhibitory activities on the TNF-R1-TNF-alpha interactions were also determined to test for specificity, and the compounds selected here all showed more than 30-fold selectivity toward the CD40-CD154 interaction. Because of their easy availability in various structural scaffolds and because of their good protein-binding ability, often explored for tissue-specific staining and other purposes, such organic dyes can provide a valuable addition to the chemical space searched to identify small molecule PPI inhibitors in general.
Collapse
Affiliation(s)
- Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA.
| | | | | | | |
Collapse
|
30
|
Lempens E, van Baal I, van Dongen J, Hackeng T, Merkx M, Meijer E. Noncovalent Synthesis of Protein Dendrimers. Chemistry 2009; 15:8760-7. [DOI: 10.1002/chem.200900748] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
31
|
Small-molecule costimulatory blockade: organic dye inhibitors of the CD40–CD154 interaction. J Mol Med (Berl) 2009; 87:1133-43. [DOI: 10.1007/s00109-009-0519-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 07/29/2009] [Accepted: 08/06/2009] [Indexed: 02/08/2023]
|
32
|
Baptiste B, Godde F, Huc I. How Can Folded Biopolymers and Synthetic Foldamers Recognize Each Other? Chembiochem 2009; 10:1765-7. [DOI: 10.1002/cbic.200900295] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
33
|
Page N, Schall N, Strub JM, Quinternet M, Chaloin O, Décossas M, Cung MT, Van Dorsselaer A, Briand JP, Muller S. The spliceosomal phosphopeptide P140 controls the lupus disease by interacting with the HSC70 protein and via a mechanism mediated by gammadelta T cells. PLoS One 2009; 4:e5273. [PMID: 19390596 PMCID: PMC2669294 DOI: 10.1371/journal.pone.0005273] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Accepted: 03/23/2009] [Indexed: 12/31/2022] Open
Abstract
The phosphopeptide P140 issued from the spliceosomal U1-70K snRNP protein is recognized by lupus CD4(+) T cells, transiently abolishes T cell reactivity to other spliceosomal peptides in P140-treated MRL/lpr mice, and ameliorates their clinical features. P140 modulates lupus patients' T cell response ex vivo and is currently included in phase IIb clinical trials. Its underlying mechanism of action remains elusive. Here we show that P140 peptide binds a unique cell-surface receptor, the constitutively-expressed chaperone HSC70 protein, known as a presenting-protein. P140 induces apoptosis of activated MRL/lpr CD4(+) T cells. In P140-treated mice, it increases peripheral blood lymphocyte apoptosis and decreases B cell, activated T cell, and CD4(-)CD8(-)B220(+) T cell counts via a specific mechanism strictly depending on gammadelta T cells. Expression of inflammation-linked genes is rapidly regulated in CD4(+) T cells. This work led us to identify a powerful pathway taken by a newly-designed therapeutic peptide to immunomodulate lupus autoimmunity.
Collapse
MESH Headings
- Animals
- Apoptosis
- B-Lymphocytes/metabolism
- Binding Sites
- Down-Regulation
- Fluorescent Antibody Technique
- HSC70 Heat-Shock Proteins/metabolism
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Lupus Erythematosus, Systemic/therapy
- Mice
- Mice, Inbred MRL lpr
- Models, Biological
- Peptide Fragments/chemistry
- Peptide Fragments/metabolism
- Peptide Fragments/pharmacology
- Receptors, Antigen, T-Cell, gamma-delta/analysis
- Ribonucleoprotein, U1 Small Nuclear/immunology
- Ribonucleoprotein, U1 Small Nuclear/metabolism
- Surface Plasmon Resonance
- T-Lymphocytes/immunology
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Nicolas Page
- CNRS UPR9021, Institut de biologie moléculaire et cellulaire, Strasbourg, France
| | - Nicolas Schall
- CNRS UPR9021, Institut de biologie moléculaire et cellulaire, Strasbourg, France
| | - Jean-Marc Strub
- CNRS UMR7178, laboratoire de spectrométrie de masse BioOrganique-IPHC-DSA- Université de Strasbourg, Strasbourg, France
| | - Marc Quinternet
- CNRS-INPL UMR7568, Laboratoire de Chimie-Physique Macromoléculaire, Nancy Université, ENSIC, Nancy, France
| | - Olivier Chaloin
- CNRS UPR9021, Institut de biologie moléculaire et cellulaire, Strasbourg, France
| | - Marion Décossas
- CNRS UPR9021, Institut de biologie moléculaire et cellulaire, Strasbourg, France
| | - Manh Thong Cung
- CNRS-INPL UMR7568, Laboratoire de Chimie-Physique Macromoléculaire, Nancy Université, ENSIC, Nancy, France
| | - Alain Van Dorsselaer
- CNRS UMR7178, laboratoire de spectrométrie de masse BioOrganique-IPHC-DSA- Université de Strasbourg, Strasbourg, France
| | - Jean-Paul Briand
- CNRS UPR9021, Institut de biologie moléculaire et cellulaire, Strasbourg, France
| | - Sylviane Muller
- CNRS UPR9021, Institut de biologie moléculaire et cellulaire, Strasbourg, France
- * E-mail:
| |
Collapse
|
34
|
Orzáez M, Gortat A, Mondragón L, Pérez-Payá E. Peptides and peptide mimics as modulators of apoptotic pathways. ChemMedChem 2009; 4:146-60. [PMID: 19021159 DOI: 10.1002/cmdc.200800246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Programmed cell death is an important and stringently controlled process. Aberrancies in its control mechanisms can lead to disease; overactive apoptosis can cause neurodegenerative disorders, whereas deficient apoptotic activity can lead to cancer. Therefore, controlling apoptotic pathways with peptides is showing increasing promise as a strategy in drug development.Programmed cell death or apoptosis is a noninvasive and strictly regulated cellular process required for organism development and tissue homeostasis. Deficiencies in apoptotic pathways are the source of many diseases such as cancer, neurodegenerative and autoimmune diseases, and disorders related to an inappropriate loss of cells such as heart failure, stroke, and liver injury. Validation of the various points of intervention as targets for drug development has been the subject of a vast number of studies. Peptides are essential tools for drug discovery, as well as preclinical and pharmaceutical drug development.
Collapse
Affiliation(s)
- Mar Orzáez
- Department of Medicinal Chemistry, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | | | | | | |
Collapse
|
35
|
Margolles-Clark E, Jacques-Silva MC, Ganesan L, Umland O, Kenyon NS, Ricordi C, Berggren PO, Buchwald P. Suramin inhibits the CD40-CD154 costimulatory interaction: a possible mechanism for immunosuppressive effects. Biochem Pharmacol 2009; 77:1236-45. [PMID: 19283894 DOI: 10.1016/j.bcp.2009.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Suramin is a symmetric polysulfonated naphthylamine-benzamide urea derivative approved for the treatment of trypanosomiasis and onchocerciasis and a known P2 (ATP/UTP purine receptor) antagonist. Here, we report its ability to inhibit the important CD40-CD154 costi-mulatory interaction required for T cell activation and the development of an effective immune response. In vitro, it inhibited the binding of both human and murine CD154 (CD40L) to their receptor (CD40) even in the presence of protein-containing media and prevented the CD154-induced proliferation of human B cells as well as the corresponding increase in surface expression of CD86, CD80, CD40, and MHC class II in a concentration-dependent manner. Furthermore, in isolated human islets, it also decreased the CD154-induced release of inflammatory cytokines such as IFN-g, interleukin-6 (IL-6), and IL-8. Suramin was selected for investigation because it has been reported to be an inhibitor of the interaction of TNF-a with its receptor and CD154 is a member of the TNF-family. However, it turned out to be a considerably, about 30-fold, more effective inhibitor of the CD40-CD154 protein-protein interaction than of the corresponding TNF interaction. Its median inhibitory concentration (IC50 50 mM) is somewhat higher than for the P2-receptor, but well within the range of its therapeutic concentration levels. Suramin shows considerable polypharmacology, but its interference with the positive costimulatory interaction might provide a possible, not yet identified mechanism for its ability to suppress T cell activity and induce immunosuppression, which might also have limited its clinical usefulness in the treatment of AIDS and cancer.
Collapse
|
36
|
Humblet V, Misra P, Bhushan KR, Nasr K, Ko YS, Tsukamoto T, Pannier N, Frangioni JV, Maison W. Multivalent scaffolds for affinity maturation of small molecule cell surface binders and their application to prostate tumor targeting. J Med Chem 2009; 52:544-50. [PMID: 19108655 DOI: 10.1021/jm801033c] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Adamantane scaffolds for affinity maturation of prostate cancer specific ligands of low molecular mass are described. These scaffolds are modular and can be used for conjugation of up to three ligands and an additional effector molecule by standard peptide coupling techniques. The potential of the scaffolds is demonstrated with the multimerization of GPI 1, a prostate cancer specific small molecule. A detailed study of multimerized GPI conjugates with near-infrared fluorophores and their binding properties to different prostate cancer cell lines shows the specific binding of these conjugates to cell types positive for prostate specific membrane antigen (PSMA). We demonstrate that these conjugates allow the sensitive imaging of prostate cancer cells with NIR methodology and suggest that our adamantane scaffolds might be generally useful for affinity maturation of small molecules targeting cell surface epitopes.
Collapse
Affiliation(s)
- Valerie Humblet
- Institute of Organic Chemistry, Justus-Liebig-Universitat Giessen, Heinrich-Buff-Ring 58, 35390 Giessen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Rationally-designed Multivalent Architectures for Mimicking Homotrimers of CD40L, a Member of the TNF Superfamily. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 611:355-7. [DOI: 10.1007/978-0-387-73657-0_157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
38
|
Law CL, Grewal IS. Therapeutic interventions targeting CD40L (CD154) and CD40: the opportunities and challenges. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 647:8-36. [PMID: 19760064 DOI: 10.1007/978-0-387-89520-8_2] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CD40 was originally identified as a receptor on B-cells that delivers contact-dependent T helper signals to B-cells through interaction with CD40 ligand (CD40L, CD154). The pivotal role played by CD40-CD40L interaction is illustrated by the defects in B-lineage cell development and the altered structures of secondary lymphoid tissues in patients and engineered mice deficient in CD40 or CD40L. CD40 signaling also provides critical functions in stimulating antigen presentation, priming of helper and cytotoxic T-cells and a variety of inflammatory reactions. As such, dysregulations in the CD40-CD40L costimulation pathway are prominently featured in human diseases ranging from inflammatory conditions to systemic autoimmunity and tissue-specific autoimmune diseases. Moreover, studies in CD40-expressing cancers have provided convincing evidence that the CD40-CD40L pathway regulates survival of neoplastic cells as well as presentation of tumor-associated antigens to the immune system. Extensive research has been devoted to explore CD40 and CD40L as drug targets. A number of anti-CD40L and anti-CD40 antibodies with diverse biological effects are in clinical development for treatment of cancer and autoimmune diseases. This chapter reviews the role of CD40-CD40L costimulation in disease pathogenesis, the characteristics of therapeutic agents targeting this pathway and status of their clinical development.
Collapse
Affiliation(s)
- Che-Leung Law
- Department of Preclinical Therapeutics, Seattle Genetics Inc., 21823 30th Drive SE, Bothell, Washington, 98021, USA.
| | | |
Collapse
|
39
|
Assembling OX40 aptamers on a molecular scaffold to create a receptor-activating aptamer. ACTA ACUST UNITED AC 2008; 15:675-82. [PMID: 18635004 DOI: 10.1016/j.chembiol.2008.05.016] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Revised: 05/16/2008] [Accepted: 05/28/2008] [Indexed: 01/31/2023]
Abstract
We show that a molecular scaffold can be utilized to convert a receptor binding aptamer into a receptor agonist. Many receptors (including tumor necrosis receptor family members) are activated when they are multimerized on the cell surface. Molecular scaffolds have been utilized to assemble multiple receptor binding peptide ligands to generate activators of such receptors. We demonstrate that an RNA aptamer that recognizes OX40, a member of the tumor necrosis factor receptor superfamily, can be converted into a receptor-activating aptamer by assembling two copies on an olignucleotide-based scaffold. The OX40 receptor-activating aptamer is able to induce nuclear localization of nuclear factor-kappaB, cytokine production, and cell proliferation, as well as enhance the potency of dendritic cell-based tumor vaccines when systemically delivered to mice.
Collapse
|
40
|
Abstract
For more than a decade now, a search for answers to the following two questions has taken us on a new and exciting journey into the world of beta- and gamma-peptides: What happens if the oxygen atoms in a 3i-helix of a polymeric chain composed of (R)-3-hydroxybutanoic acid are replaced by NH units? What happens if one or two CH2 groups are introduced into each amino acid building block in the chain of a peptide or protein, thereby providing homologues of the proteinogenic alpha-amino acids? Our journey has repeatedly thrown up surprises, continually expanding the potential of these classes of compound and deepening our understanding of the structures, properties, and multifaceted functions of the natural "models" to which they are related. Beta-peptides differ from their natural counterparts, the alpha-peptides, by having CH2 groups inserted into every amino acid residue, either between the C=O groups and the alpha-carbon atoms (beta(3)) or between the alpha-carbon and nitrogen atoms (beta(2)). The synthesis of these homologated proteinogenic amino acids and their assembly into beta-peptides can be performed using known methods. Despite the increased number of possible conformers, the beta-peptides form secondary structures (helices, turns, sheets) even when the chain lengths are as short as four residues. Furthermore, they are stable toward degrading and metabolizing enzymes in living organisms. Linear, helical, and hairpin-type structures of beta-peptides can now be designed in such a way that they resemble the characteristic and activity-related structural features ("epitopes") of corresponding natural peptides or protein sections. This Account presents examples of beta-peptidic compounds binding, as agonists or antagonists (inhibitors), to (i) major histocompatibility complex (MHC) proteins (immune response), (ii) the lipid-transport protein SR-B1 (cholesterol uptake from the small intestine), (iii) the core (1-60) of interleukin-8 (inflammation), (iv) the oncoprotein RDM2, (v) the HIVgp41 fusion protein, (vi) G-protein-coupled somatostatin hsst receptors, (vii) the TNF immune response receptor CD40 (apoptosis), and (viii) DNA. Short-chain beta-peptides may be orally bioavailable and excreted from the body of mammals; long-chain beta-peptides may require intravenous administration but will have longer half-lives of clearance. It has been said that an interesting field of research distinguishes itself in that the results always throw up new questions; in this sense, the structural and biological investigation of beta-peptides has been a gold mine. We expect that these peptidic peptidomimetics will play an increasing role in biomedical research and drug development in the near future.
Collapse
Affiliation(s)
- Dieter Seebach
- Laboratorium für Organische Chemie, Departement Chemie und Angewandte Biowissenschaften, ETH Zürich, Wolfgang-Pauli-Strasse 10, CH-8093 Zürich, Switzerland
| | - James Gardiner
- Laboratorium für Organische Chemie, Departement Chemie und Angewandte Biowissenschaften, ETH Zürich, Wolfgang-Pauli-Strasse 10, CH-8093 Zürich, Switzerland
| |
Collapse
|
41
|
Seebach D, Dubost E, Mathad R, Jaun B, Limbach M, Löweneck M, Flögel O, Gardiner J, Capone S, Beck A, Widmer H, Langenegger D, Monna D, Hoyer D. New Open-Chain and Cyclic Tetrapeptides, Consisting ofα-,β2-, andβ3-Amino-Acid Residues, as Somatostatin Mimics - A Survey. Helv Chim Acta 2008. [DOI: 10.1002/hlca.200890190] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
42
|
|
43
|
Le Grel P, Salaün A, Mocquet C, Le Grel B, Roisnel T, Potel M. Postsynthetic Modification of C3-Symmetric Aza-β3-Cyclohexapeptides. J Org Chem 2008; 73:1306-10. [DOI: 10.1021/jo7021394] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Philippe Le Grel
- ICMV and CSM, UMR CNRS 6226, Université de Rennes I, 263 avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Arnaud Salaün
- ICMV and CSM, UMR CNRS 6226, Université de Rennes I, 263 avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Clémence Mocquet
- ICMV and CSM, UMR CNRS 6226, Université de Rennes I, 263 avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Barbara Le Grel
- ICMV and CSM, UMR CNRS 6226, Université de Rennes I, 263 avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Thierry Roisnel
- ICMV and CSM, UMR CNRS 6226, Université de Rennes I, 263 avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Michel Potel
- ICMV and CSM, UMR CNRS 6226, Université de Rennes I, 263 avenue du Général Leclerc, 35042 Rennes Cedex, France
| |
Collapse
|
44
|
Nasr K, Pannier N, Frangioni JV, Maison W. Rigid multivalent scaffolds based on adamantane. J Org Chem 2008; 73:1056-60. [PMID: 18179237 DOI: 10.1021/jo702310g] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We present two new synthetic strategies to rigid multivalent scaffolds of the general structure 1 based on adamantane. Both routes start from arylated adamantane derivatives and give the target compounds 12 and 18 in 5 and 7 steps, respectively. These scaffolds have been designed for the assembly of multivalent binders for cell surface epitopes. The adamantane nucleus exposes three carboxylic acid groups in a well-defined tripodal geometry for conjugation of targeting ligands. In addition, an amino group at the fourth bridgehead position provides a flexible linker for attachment of effector molecules such as contrast agents, radiotracers, or cytotoxins without interfering with the cell binding process.
Collapse
Affiliation(s)
- Khaled Nasr
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, SL-B05, 330 Brookline Avenue, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
45
|
Zapata JM, Lefebvre S, Reed JC. Targeting TRAFs for Therapeutic Intervention. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 597:188-201. [PMID: 17633027 DOI: 10.1007/978-0-387-70630-6_15] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
TNF-receptor associated factors (TRAFs) are the molecules that upon engagement of the TNF-receptor (TNFR) by a TNF-family ligand come first in contact with the activated TNFR, initially acting as docking molecules for kinases and other effector proteins that are recruited to the activated receptor. TRAFs later regulate the subcellular relocalization of the receptor-ligand complex and finally they modulate the extent of the response by controlling the degradation of key proteins in the pathway. In this chapter, we review the involvement of different TRAF family members in the etiology of a variety of pathologies and address the question of whether the use of TNFR-mimic-peptides or small molecule modulators targeting TRAFs might be suitable for therapeutic intervention, discussing the advantages and disadvantages of this strategy.
Collapse
Affiliation(s)
- Juan M Zapata
- Burnham Institute for Medical Research, La Jolla, California 92037, USA.
| | | | | |
Collapse
|
46
|
Holub J, Garabedian M, Kirshenbaum K. Peptoids on Steroids: Precise Multivalent Estradiol-Peptidomimetic Conjugates Generated via
Azide-Alkyne [3+2] Cycloaddition Reactions. ACTA ACUST UNITED AC 2007. [DOI: 10.1002/qsar.200740074] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
47
|
Trouche N, Wieckowski S, Sun W, Chaloin O, Hoebeke J, Fournel S, Guichard G. Small Multivalent Architectures Mimicking Homotrimers of the TNF Superfamily Member CD40L: Delineating the Relationship between Structure and Effector Function. J Am Chem Soc 2007; 129:13480-92. [DOI: 10.1021/ja073169m] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Nathalie Trouche
- Contribution from the CNRS, Institut de Biologie Moléculaire et Cellulaire, laboratoire d'Immunologie et Chimie Thérapeutiques, 15 rue René Descartes, 67084 Strasbourg, France
| | - Sébastien Wieckowski
- Contribution from the CNRS, Institut de Biologie Moléculaire et Cellulaire, laboratoire d'Immunologie et Chimie Thérapeutiques, 15 rue René Descartes, 67084 Strasbourg, France
| | - Weimin Sun
- Contribution from the CNRS, Institut de Biologie Moléculaire et Cellulaire, laboratoire d'Immunologie et Chimie Thérapeutiques, 15 rue René Descartes, 67084 Strasbourg, France
| | - Olivier Chaloin
- Contribution from the CNRS, Institut de Biologie Moléculaire et Cellulaire, laboratoire d'Immunologie et Chimie Thérapeutiques, 15 rue René Descartes, 67084 Strasbourg, France
| | - Johan Hoebeke
- Contribution from the CNRS, Institut de Biologie Moléculaire et Cellulaire, laboratoire d'Immunologie et Chimie Thérapeutiques, 15 rue René Descartes, 67084 Strasbourg, France
| | - Sylvie Fournel
- Contribution from the CNRS, Institut de Biologie Moléculaire et Cellulaire, laboratoire d'Immunologie et Chimie Thérapeutiques, 15 rue René Descartes, 67084 Strasbourg, France
| | - Gilles Guichard
- Contribution from the CNRS, Institut de Biologie Moléculaire et Cellulaire, laboratoire d'Immunologie et Chimie Thérapeutiques, 15 rue René Descartes, 67084 Strasbourg, France
| |
Collapse
|
48
|
Habib M, Noval Rivas M, Chamekh M, Wieckowski S, Sun W, Bianco A, Trouche N, Chaloin O, Dumortier H, Goldman M, Guichard G, Fournel S, Vray B. Cutting edge: small molecule CD40 ligand mimetics promote control of parasitemia and enhance T cells producing IFN-gamma during experimental Trypanosoma cruzi infection. THE JOURNAL OF IMMUNOLOGY 2007; 178:6700-4. [PMID: 17513713 DOI: 10.4049/jimmunol.178.11.6700] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Host resistance to Trypanosoma cruzi infection depends on a type 1 response characterized by a strong production of IL-12 and IFN-gamma. Amplifying this response through CD40 triggering results in control of parasitemia. Two newly synthesized molecules (<3 kDa) mimicking trimeric CD40L (mini CD40Ls(-1) and (-2)) bind to CD40, activate murine dendritic cells, and elicit IL-12 production. Wild-type but not CD40 knockout mice exhibited a sharp decrease of parasitemia and mortality when inoculated with T. cruzi mixed with miniCD40Ls. Moreover, the immunosuppression induced by T. cruzi infection was impaired in mice treated with miniCD40Ls, as shown by proliferation of splenic lymphocytes, percentage of CD8(+) T cells, and IFN-gamma production. Mice surviving T. cruzi infection in the presence of miniCD40L(-1) were immunized against a challenge infection. Our results indicate that CD40L mimetics are effective in vivo and promote the control of T. cruzi infection by overcoming the immunosuppression usually induced by the parasites.
Collapse
Affiliation(s)
- Mohammed Habib
- Laboratoire d'Immunologie Expérimentale, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Affiliation(s)
- Susan E Gibson
- Department of Chemistry, Imperial College London, South Kensington Campus, London SW7 2AY, UK.
| | | |
Collapse
|
50
|
|