1
|
Belsuzarri MM, Sako Y, Brown TD, Chan M, Cozza R, Jin J, Sato-Kaneko F, Yao S, Pu M, Messer K, Hayashi T, Cottam HB, Corr M, Carson DA, Shukla NM. Structure-Activity Relationship Studies in Benzothiadiazoles as Novel Vaccine Adjuvants. J Med Chem 2024; 67:13703-13722. [PMID: 39115891 DOI: 10.1021/acs.jmedchem.4c00491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Extracellular vesicles (EVs) can transfer antigens and immunomodulatory molecules, and such EVs released by antigen-presenting cells equipped with immunostimulatory functions have been utilized for vaccine formulations. A prior high-throughput screening campaign led to the identification of compound 634 (1), which enhanced EV release and increased intracellular Ca2+ influx. Here, we performed systematic structure-activity relationship (SAR) studies to investigate the scaffold for its potency as a vaccine adjuvant. Synthesized compounds were analyzed in vitro for CD63 reporter activity (a marker for EV biogenesis) in human THP-1 cells, induction of Ca2+ influx, IL-12 production, and cell viability in murine bone-marrow-derived dendritic cells. The SAR studies indicated that the ester functional group was requisite, and the sulfur atom of the benzothiadiazole ring replaced with a higher selenium atom (9f) or a bioisosteric ethenyl group (9h) retained potency. Proof-of-concept vaccination studies validated the potency of the selected compounds as novel vaccine adjuvants.
Collapse
Affiliation(s)
- Masiel M Belsuzarri
- UC San Diego Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Yukiya Sako
- UC San Diego Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Tyler D Brown
- UC San Diego Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Michael Chan
- UC San Diego Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Renna Cozza
- UC San Diego Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Jasmine Jin
- UC San Diego Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Fumi Sato-Kaneko
- Department of Medicine, University of California San Diego, La Jolla, California 92093, United States
| | - Shiyin Yao
- UC San Diego Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Minya Pu
- UC San Diego Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Karen Messer
- Division of Biostatistics, Herbert Wertheim School of Public Health and Longevity, University of California San Diego, La Jolla, California 92093, United States
| | - Tomoko Hayashi
- Department of Medicine, University of California San Diego, La Jolla, California 92093, United States
| | - Howard B Cottam
- Department of Medicine, University of California San Diego, La Jolla, California 92093, United States
| | - Maripat Corr
- Department of Medicine, University of California San Diego, La Jolla, California 92093, United States
| | - Dennis A Carson
- Department of Medicine, University of California San Diego, La Jolla, California 92093, United States
| | - Nikunj M Shukla
- Department of Medicine, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
2
|
Yu B, Li B, Chen T, Yang J, Wang X, Peng B, Hu Q. A NF-κB-Based High-Throughput Screening for Immune Adjuvants and Inhibitors. Inflammation 2023; 46:598-611. [PMID: 36306023 DOI: 10.1007/s10753-022-01758-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/19/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022]
Abstract
The nuclear factor-κB (NF-κB) family is crucial for regulating immune and inflammatory responses. The activation of the immune cell signaling pathway usually activates NF-κB, causing a protective immune response. NF-κB can also cause excessive inflammatory responses by activating a cascade reaction of pro-inflammatory mediators such as cytokines. In this study, we used an NF-κB luciferase reporter gene system. Out of more than 800 compounds screened, four NF-κB agonists were identified with strong activity at nontoxic concentrations. Subsequently, the adjuvant effect was verified on mouse bone marrow-derived dendritic cells (BMDCs) and macrophages RAW264.7. It was found that fostamatinib (R788) disodium increased the production of IL-6, IL-12p40, and TNF-α, indicating that R788 disodium could induce the maturation of antigen-presenting cells (APCs). In addition, three compounds were screened to significantly inhibit NF-κB at nontoxic doses, including dehydrocostus lactone (DHL)-a known NF-κB inhibitor. The results showed that DHL significantly reduced the release of LPS-induced inflammatory cytokines (including TNF-α, IL-6, and IL-12). Our findings indicate that the NF-κB-based high-throughput screening can be used to discover potential immune adjuvants and anti-inflammatory molecules.
Collapse
Affiliation(s)
- Boyang Yu
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Boye Li
- Civil Aviation Medicine Center, Civil Aviation Administration of China, Beijing, 100123, China
| | - Tian Chen
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Jinning Yang
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Xiaoli Wang
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Bo Peng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Qin Hu
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China.
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China.
| |
Collapse
|
3
|
Zhao M, Yang K, Zhu X, Gao T, Yu W, Liu H, You Z, Liu Z, Qiao X, Song Y. Design, synthesis and biological evaluation of dual Topo II/HDAC inhibitors bearing pyrimido[5,4-b]indole and pyrazolo[3,4-d]pyrimidine motifs. Eur J Med Chem 2023; 252:115303. [PMID: 36996717 DOI: 10.1016/j.ejmech.2023.115303] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/21/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Both topoisomerase II (Topo II) and histone deacetylase (HDAC) are important therapeutic targets for cancer. In this study, two series of novel compounds containing pyrimido[5,4-b]indole and pyrazolo[3,4-d]pyrimidine motifs were designed and synthesized as dual Topo II/HDAC inhibitors. MTT assay indicated that all the compounds displayed potential antiproliferative activity against three cancer cell lines (MGC-803, MCF-7 and U937) and low cytotoxicity on normal cell line (3T3). In the enzyme activity inhibition experiments, compounds 7d and 8d exhibited excellent dual inhibitory activities against Topo II and HDAC. Cleavage reaction assay showed that 7d was a Topo II poison, which was consistent with the docking results. Further experimental results revealed that compounds 7d and 8d could promote apoptosis and significantly inhibit the migration in MCF-7 cells. Molecular docking showed that compounds 7d and 8d bind Topo II and HDAC at the active sites. Molecular dynamics simulation showed that 7d can stably bind to Topo II and HDAC.
Collapse
Affiliation(s)
- Mengmiao Zhao
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei, 071002, China
| | - Kan Yang
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei, 071002, China
| | - Xinyue Zhu
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei, 071002, China
| | - Tian Gao
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei, 071002, China
| | - Wei Yu
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei, 071002, China
| | - Han Liu
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei, 071002, China
| | - Zhihao You
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei, 071002, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xiaoqiang Qiao
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei, 071002, China; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education, Hebei University, Baoding, Hebei, 071002, China.
| | - Yali Song
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei, 071002, China; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education, Hebei University, Baoding, Hebei, 071002, China.
| |
Collapse
|
4
|
Devasia J, Nizam A, V. L. V. Azole-Based Antibacterial Agents: A Review on Multistep Synthesis Strategies and Biology. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2021.1938615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Jyothis Devasia
- Department of Chemistry, CHRIST (Deemed to be University), Bangalore, Karnataka, India
| | - Aatika Nizam
- Department of Chemistry, CHRIST (Deemed to be University), Bangalore, Karnataka, India
| | - Vasantha V. L.
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, Karnataka, India
| |
Collapse
|
5
|
Wu Q, Xu G, Li W, Wan J, Liu T, Huang C. Cu(I)-Catalyzed and Base-Promoted [5 + 2 + 1] Cascade Cyclization of 2-Nitrochalcones with Aliphatic Primary Amines to 5 H-Pyrimido[5,4- b]indole Frameworks. Org Lett 2022; 24:3950-3954. [PMID: 35639963 DOI: 10.1021/acs.orglett.2c01289] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
An unprecedented [5 + 2 + 1] cascade cyclization to the preparation of 5H-pyrimido[5,4-b]indole derivatives has been disclosed. The novel protocol of 2-nitrochalcones reacted with aliphatic primary amines catalyzed by CuI and promoted by Cs2CO3, which underwent a critical intermediate 2,3-disubstituted indole, providing structurally diverse 5H-pyrimido[5,4-b]indoles in generally high yields (77-90%) and broad substrate scopes (34 examples).
Collapse
Affiliation(s)
- Qin Wu
- National and Local Joint Engineering Research Center for Green Preparation Technology of Biobased Materials, School of Chemistry and Environment, Yunnan Minzu University, Kunming 650500, P. R. China
| | - Genrui Xu
- National and Local Joint Engineering Research Center for Green Preparation Technology of Biobased Materials, School of Chemistry and Environment, Yunnan Minzu University, Kunming 650500, P. R. China
| | - Weiqiang Li
- National and Local Joint Engineering Research Center for Green Preparation Technology of Biobased Materials, School of Chemistry and Environment, Yunnan Minzu University, Kunming 650500, P. R. China
| | - Juan Wan
- National and Local Joint Engineering Research Center for Green Preparation Technology of Biobased Materials, School of Chemistry and Environment, Yunnan Minzu University, Kunming 650500, P. R. China
| | - Teng Liu
- College of Chemistry and Environmental Science, Qujing Normal University, Qujing 655011, P. R. China
| | - Chao Huang
- National and Local Joint Engineering Research Center for Green Preparation Technology of Biobased Materials, School of Chemistry and Environment, Yunnan Minzu University, Kunming 650500, P. R. China
| |
Collapse
|
6
|
Shukla NM, Sato-Kaneko F, Yao S, Pu M, Chan M, Lao FS, Sako Y, Saito T, Messer K, Hayashi T, Cottam HB, Corr M, Carson DA. A Triple High Throughput Screening for Extracellular Vesicle Inducing Agents With Immunostimulatory Activity. Front Pharmacol 2022; 13:869649. [PMID: 35479316 PMCID: PMC9035538 DOI: 10.3389/fphar.2022.869649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
Extracellular vesicles (EVs) play an important role in intercellular communication and regulation of cells, especially in the immune system where EVs can participate in antigen presentation and may have adjuvant effects. We aimed to identify small molecule compounds that can increase EV release and thereby enhance the immunogenicity of vaccines. We utilized a THP-1 reporter cell line engineered to release EV-associated tetraspanin (CD63)-Turbo-luciferase to quantitatively measure EVs released in culture supernatants as a readout of a high throughput screen (HTS) of 27,895 compounds. In parallel, the cytotoxicity of the compounds was evaluated by PrestoBlue dye assay. For screening immunostimulatory potency, we performed two additional independent HTS on the same compound library using NF-κB and interferon-stimulated response element THP-1 reporter cell lines. Hit compounds were then identified in each of the 3 HTS’s, using a “Top X″ and a Gaussian Mixture Model approach to rule out false positive compounds and to increase the sensitivity of the hit selection. Thus, 644 compounds were selected as hits which were further evaluated for induction of IL-12 in murine bone-marrow derived dendritic cells (mBMDCs) and for effects of cell viability. The resulting 130 hits were then assessed from a medicinal chemistry perspective to remove compounds with functional group liabilities. Finally, 80 compounds were evaluated as vaccine adjuvants in vivo using ovalbumin as a model antigen. We analyzed 18 compounds with adjuvant activity for their ability to induce the expression of co-stimulatory molecules on mBMDCs. The full complement of data was then used to cluster the compounds into 4 distinct biological activity profiles. These compounds were also evaluated for quantitation of EV release and spider plot overlays were generated to compare the activity profiles of compounds within each cluster. This tiered screening process identified two compounds that belong to the 4-thieno-2-thiopyrimidine scaffold with identical screening profiles supporting data reproducibility and validating the overall screening process. Correlation patterns in the adjuvanticity data suggested a role for CD63 and NF-κB pathways in potentiating antigen-specific antibody production. Thus, our three independent cell-based HTS campaigns led to identification of immunostimulatory compounds that release EVs and have adjuvant activity.
Collapse
Affiliation(s)
- Nikunj M. Shukla
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Fumi Sato-Kaneko
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Shiyin Yao
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Minya Pu
- The Herbert Wertheim School of Public Health and Longevity, University of California San Diego, La Jolla, CA, United States
| | - Michael Chan
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Fitzgerald S. Lao
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Yukiya Sako
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Tetsuya Saito
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Karen Messer
- The Herbert Wertheim School of Public Health and Longevity, University of California San Diego, La Jolla, CA, United States
| | - Tomoko Hayashi
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Howard B. Cottam
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Maripat Corr
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Dennis A. Carson
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
- *Correspondence: Dennis A. Carson,
| |
Collapse
|
7
|
Saito T, Shukla NM, Sato-Kaneko F, Sako Y, Hosoya T, Yao S, Lao FS, Messer K, Pu M, Chan M, Chu PJ, Cottam HB, Hayashi T, Carson DA, Corr M. Small Molecule Calcium Channel Activator Potentiates Adjuvant Activity. ACS Chem Biol 2022; 17:217-229. [PMID: 34985883 PMCID: PMC8788586 DOI: 10.1021/acschembio.1c00883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/21/2021] [Indexed: 01/07/2023]
Abstract
There remains an unmet need for reliable fully synthetic adjuvants that increase lasting protective immune responses from vaccines. We previously reported a high-throughput screening for small molecules that extended nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) activation after a Toll-like receptor 4 (TLR4) ligand, lipopolysaccharide (LPS), stimulation using a human myeloid reporter cell line. We identified compounds with a conserved aminothiazole scaffold including 2D216 [N-(4-(2,5-dimethylphenyl)thiazol-2-yl)-4-(piperidin-1-ylsulfonyl)benzamide], which increased murine antigen-specific antibody responses when used as a co-adjuvant with LPS. Here, we examined the mechanism of action in human cells. Although 2D216 activated the major mitogen-activated protein kinases, it did not interact with common kinases and phosphatases and did not stimulate many of the pattern recognition receptors (PRRs). Instead, the mechanism of action was linked to intracellular Ca2+ elevation via Ca2+ channel(s) at the plasma membrane and nuclear translocation of the nuclear factor of activated T-cells (NFAT) as supported by RNA-seq data, analysis by reporter cells, Ca2+ flux assays, and immunoblots. Interestingly, 2D216 had minimal, if any, activity on Jurkat T cells but induced cytokine production and surface expression of costimulatory molecules on cells with antigen-presenting functions. A small series of analogs of 2D216 were tested for the ability to enhance a TLR4 ligand-stimulated autologous mixed lymphocyte reaction (MLR). In the MLR, 2E151, N-(4-(2,5-dimethylphenyl)thiazol-2-yl)-4-((4-propylpiperidin-1-yl)sulfonyl)benzamide, was more potent than 2D216. These results indicate that a small molecule that is not a direct PRR agonist can act as a co-adjuvant to an approved adjuvant to enhance human immune responses via a complementary mechanism of action.
Collapse
Affiliation(s)
- Tetsuya Saito
- Moores
Cancer Center, University of California
San Diego, La Jolla, California 92093-0809, United States
- Department
of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8519, Japan
| | - Nikunj M. Shukla
- Moores
Cancer Center, University of California
San Diego, La Jolla, California 92093-0809, United States
| | - Fumi Sato-Kaneko
- Moores
Cancer Center, University of California
San Diego, La Jolla, California 92093-0809, United States
| | - Yukiya Sako
- Moores
Cancer Center, University of California
San Diego, La Jolla, California 92093-0809, United States
| | - Tadashi Hosoya
- Moores
Cancer Center, University of California
San Diego, La Jolla, California 92093-0809, United States
- Department
of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8519, Japan
| | - Shiyin Yao
- Moores
Cancer Center, University of California
San Diego, La Jolla, California 92093-0809, United States
| | - Fitzgerald S. Lao
- Moores
Cancer Center, University of California
San Diego, La Jolla, California 92093-0809, United States
| | - Karen Messer
- Herbert
Wertheim School of Public Health and Longevity, University of California San Diego, La Jolla, California 92093-0901, United States
| | - Minya Pu
- Herbert
Wertheim School of Public Health and Longevity, University of California San Diego, La Jolla, California 92093-0901, United States
| | - Michael Chan
- Moores
Cancer Center, University of California
San Diego, La Jolla, California 92093-0809, United States
| | - Paul J. Chu
- Moores
Cancer Center, University of California
San Diego, La Jolla, California 92093-0809, United States
| | - Howard B. Cottam
- Moores
Cancer Center, University of California
San Diego, La Jolla, California 92093-0809, United States
| | - Tomoko Hayashi
- Moores
Cancer Center, University of California
San Diego, La Jolla, California 92093-0809, United States
| | - Dennis A. Carson
- Moores
Cancer Center, University of California
San Diego, La Jolla, California 92093-0809, United States
| | - Maripat Corr
- Department
of Medicine, University of California San
Diego, La Jolla, California 92093-0656, United States
| |
Collapse
|
8
|
Saito T, Sako Y, Sato-Kaneko F, Hosoya T, Yao S, Lao FS, Shpigelman J, Messer K, Pu M, Shukla NM, Chan M, Chu PJ, Cottam HB, Hayashi T, Carson DA, Corr M. Small Molecule Potentiator of Adjuvant Activity Enhancing Survival to Influenza Viral Challenge. Front Immunol 2021; 12:701445. [PMID: 34650551 PMCID: PMC8505803 DOI: 10.3389/fimmu.2021.701445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/30/2021] [Indexed: 01/02/2023] Open
Abstract
As viruses continue to mutate the need for rapid high titer neutralizing antibody responses has been highlighted. To meet these emerging threats, agents that enhance vaccine adjuvant activity are needed that are safe with minimal local or systemic side effects. To respond to this demand, we sought small molecules that would sustain and improve the protective effect of a currently approved adjuvant, monophosphoryl lipid A (MPLA), a Toll-like receptor 4 (TLR4) agonist. A lead molecule from a high-throughput screen, (N-(4-(2,5-dimethylphenyl)thiazol-2-yl)-4-(piperidin-1-ylsulfonyl)benzamide, was identified as a hit compound that sustained NF-κB activation by a TLR4 ligand, lipopolysaccharide (LPS), after an extended incubation (16 h). In vitro, the resynthesized compound (2D216) enhanced TLR4 ligand-induced innate immune activation and antigen presenting function in primary murine bone marrow-derived dendritic cells without direct activation of T cells. In vivo murine vaccination studies demonstrated that compound 2D216 acted as a potent co-adjuvant when used in combination with MPLA that enhanced antigen-specific IgG equivalent to that of AS01B. The combination adjuvant MPLA/2D216 produced Th1 dominant immune responses and importantly protected mice from lethal influenza virus challenge. 2D216 alone or 2D216/MPLA demonstrated minimal local reactogenicity and no systemic inflammatory response. In summary, 2D216 augmented the beneficial protective immune responses of MPLA as a co-adjuvant and showed an excellent safety profile.
Collapse
Affiliation(s)
- Tetsuya Saito
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States.,Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yukiya Sako
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Fumi Sato-Kaneko
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Tadashi Hosoya
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States.,Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shiyin Yao
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Fitzgerald S Lao
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Jonathan Shpigelman
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Karen Messer
- The Herbert Wertheim School of Public Health and Longevity, University of California San Diego, La Jolla, CA, United States
| | - Minya Pu
- The Herbert Wertheim School of Public Health and Longevity, University of California San Diego, La Jolla, CA, United States
| | - Nikunj M Shukla
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Michael Chan
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Paul J Chu
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Howard B Cottam
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Tomoko Hayashi
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Dennis A Carson
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Maripat Corr
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
9
|
Hosoya T, Cordelia D, Michael BD, Miyabe C, Nagai J, Murooka TT, Miyabe Y. Editorial: Targeting the Chemoattractant System in Inflammation. Front Pharmacol 2021; 12:744290. [PMID: 34483948 PMCID: PMC8415622 DOI: 10.3389/fphar.2021.744290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/04/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Tadashi Hosoya
- Department of Rheumatology, Tokyo Medical and Dental University (TMDU), Liverpool, United Kingdom
| | - Dunai Cordelia
- Clinical Infection Microbiology and Immunology, Institute of Infection Ecology and Veterinary Sciences, University of Liverpool, Liverpool, United Kingdom.,NIHR HPRU for Emerging and Zoonotic Infection, Liverpool, United Kingdom
| | - Benedict D Michael
- Clinical Infection Microbiology and Immunology, Institute of Infection Ecology and Veterinary Sciences, University of Liverpool, Liverpool, United Kingdom.,NIHR HPRU for Emerging and Zoonotic Infection, Liverpool, United Kingdom.,The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
| | - Chie Miyabe
- Division of Dermatology, Tokyo Women's Medical University, Chiba, Japan
| | - Jun Nagai
- Department of Medicine, Harvard Medical School, Boston, MA, United States.,Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, United States
| | - Thomas T Murooka
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Yoshishige Miyabe
- Department of Cell Biology, Nippon Medical School, Institute for Advanced Medical Sciences, Tokyo, Japan
| |
Collapse
|
10
|
Shukla NM, Chan M, Lao FS, Chu PJ, Belsuzarri M, Yao S, Nan J, Sato-Kaneko F, Saito T, Hayashi T, Corr M, Carson DA, Cottam HB. Structure-activity relationship studies in substituted sulfamoyl benzamidothiazoles that prolong NF-κB activation. Bioorg Med Chem 2021; 43:116242. [PMID: 34274759 DOI: 10.1016/j.bmc.2021.116242] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/19/2022]
Abstract
In the face of emerging infectious diseases, there remains an unmet need for vaccine development where adjuvants that enhance immune responses to pathogenic antigens are highly desired. Using high-throughput screens with a cell-based nuclear factor κB (NF-κB) reporter assay, we identified a sulfamoyl benzamidothiazole bearing compound 1 that demonstrated a sustained activation of NF-κB after a primary stimulus with a Toll-like receptor (TLR)-4 agonist, lipopolysaccharide (LPS). Here, we explore systematic structure-activity relationship (SAR) studies on compound 1 that indicated the sites on the scaffold that tolerated modification and yielded more potent compounds compared to 1. The selected analogs enhanced release of immunostimulatory cytokines in the human monocytic cell line THP-1 cells and murine primary dendritic cells. In murine vaccination studies, select compounds were used as co-adjuvants in combination with the Food and Drug Administration approved TLR-4 agonistic adjuvant, monophosphoryl lipid A (MPLA) that showed significant enhancement in antigen-specific antibody titers compared to MPLA alone. Additionally, our SAR studies led to identification of a photoaffinity probe which will aid the target identification and mechanism of action studies in the future.
Collapse
Affiliation(s)
- Nikunj M Shukla
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0809, USA.
| | - Michael Chan
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0809, USA
| | - Fitzgerald S Lao
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0809, USA
| | - Paul J Chu
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0809, USA
| | - Masiel Belsuzarri
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0809, USA
| | - Shiyin Yao
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0809, USA
| | - Jason Nan
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0809, USA
| | - Fumi Sato-Kaneko
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0809, USA
| | - Tetsuya Saito
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0809, USA
| | - Tomoko Hayashi
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0809, USA
| | - Maripat Corr
- Department of Medicine, University of California San Diego, La Jolla, CA 92093-0656, USA
| | - Dennis A Carson
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0809, USA
| | - Howard B Cottam
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0809, USA
| |
Collapse
|
11
|
Hosoya T, Shukla NM, Fujita Y, Yao S, Lao FS, Baba H, Yasuda S, Cottam HB, Carson DA, Hayashi T, Corr M. Identification of Compounds With Glucocorticoid Sparing Effects on Suppression of Chemokine and Cytokine Production by Rheumatoid Arthritis Fibroblast-Like Synoviocytes. Front Pharmacol 2020; 11:607713. [PMID: 33390996 PMCID: PMC7773657 DOI: 10.3389/fphar.2020.607713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/04/2020] [Indexed: 12/19/2022] Open
Abstract
In recent years target based drug discovery has expanded our therapeutic armamentarium in the treatment of inflammatory and autoimmune diseases. Despite these advances and adverse effects, glucocorticoids remain reliable agents that are used in many of these diseases. The anti-inflammatory mechanisms of glucocorticoids include the suppression of transcription factor activity like nuclear factor kappa B (NF-κB). By reanalyzing data from two prior high throughput screens (HTS) that utilized a NF-κB reporter construct in THP-1 cells, we identified 1824 small molecule synthetic compounds that demonstrated NF-κB suppressive activities similar to the glucocorticoids included in the original >134,000 compound libraries. These 1824 compounds were then rescreened for attenuating NF-κB activity at 5 and 16 h after LPS stimuli in the NF-κB THP-1 reporter cells. After a “Top X” selection approach 122 hit compounds were further tested for toxicity and suppression of LPS induced CXCL8 release in THP-1 cells. Excluding cytotoxic compounds, the remaining active compounds were grouped into chemotype families using Tanimoto based clustering. Promising representatives from clustered chemotype groups were commercially purchased for further testing. Amongst these index compounds a lead chemotype: 1H-pyrazolo [3,4 d] pyrimidin-4-amine, effectively suppressed CXCL8, and TNF production by THP-1 cells when stimulated with LPS, TNF or IL-1ß. Extending these studies to primary cells, these lead compounds also reduced IL-6 and CXCL8 production by TNF stimulated fibroblast-like synoviocytes (FLS) from rheumatoid arthritis (RA) patients. Importantly a lead 1H-pyrazolo [3,4 d] pyrimidin-4-amine compound demonstrated synergistic effects with dexamethasone when co-administered to TNF stimulated THP-1 cells and RA FLS in suppressing chemokine production. In summary, a cell based HTS approach identified lead compounds that reduced NF-κB activity and chemokine secretion induced by potent immunologic stimuli, and one lead compound that acted synergistically with dexamethasone as an anti-inflammatory agent showing a dose-sparing effect.
Collapse
Affiliation(s)
- Tadashi Hosoya
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States.,Department of Rheumatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Nikunj M Shukla
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Yuya Fujita
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Shiyin Yao
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Fitzgerald S Lao
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Hiroyuki Baba
- Department of Rheumatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinsuke Yasuda
- Department of Rheumatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Howard B Cottam
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Dennis A Carson
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Tomoko Hayashi
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Maripat Corr
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
12
|
Van Herck S, De Geest BG. Nanomedicine-mediated alteration of the pharmacokinetic profile of small molecule cancer immunotherapeutics. Acta Pharmacol Sin 2020; 41:881-894. [PMID: 32451411 PMCID: PMC7471422 DOI: 10.1038/s41401-020-0425-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/20/2020] [Indexed: 12/21/2022]
Abstract
The advent of immunotherapy is a game changer in cancer therapy with monoclonal antibody- and T cell-based therapeutics being the current flagships. Small molecule immunotherapeutics might offer advantages over the biological drugs in terms of complexity, tissue penetration, manufacturing cost, stability, and shelf life. However, small molecule drugs are prone to rapid systemic distribution, which might induce severe off-target side effects. Nanotechnology could aid in the formulation of the drug molecules to improve their delivery to specific immune cell subsets. In this review we summarize the current efforts in changing the pharmacokinetic profile of small molecule immunotherapeutics with a strong focus on Toll-like receptor agonists. In addition, we give our vision on limitations and future pathways in the route of nanomedicine to the clinical practice.
Collapse
Affiliation(s)
- Simon Van Herck
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Bruno G De Geest
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium.
| |
Collapse
|
13
|
Chan M, Lao FS, Chu PJ, Shpigelman J, Yao S, Nan J, Sato-Kaneko F, Li V, Hayashi T, Corr M, Carson DA, Cottam HB, Shukla NM. Structure–Activity Relationship Studies To Identify Affinity Probes in Bis-aryl Sulfonamides That Prolong Immune Stimuli. J Med Chem 2019; 62:9521-9540. [DOI: 10.1021/acs.jmedchem.9b00870] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Michael Chan
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0695, United States
| | - Fitzgerald S. Lao
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0695, United States
| | - Paul J. Chu
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0695, United States
| | - Jonathan Shpigelman
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0695, United States
| | - Shiyin Yao
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0695, United States
| | - Jason Nan
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0695, United States
| | - Fumi Sato-Kaneko
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0695, United States
| | - Vicky Li
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0695, United States
| | - Tomoko Hayashi
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0695, United States
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | - Dennis A. Carson
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0695, United States
| | - Howard B. Cottam
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0695, United States
| | - Nikunj M. Shukla
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0695, United States
| |
Collapse
|
14
|
Shukla N, Chan M, Hayashi T, Carson DA, Cottam HB. Recent Advances and Perspectives in Small-molecule TLR Ligands and Their Modulators. ACS Med Chem Lett 2018; 9:1156-1159. [PMID: 30613317 PMCID: PMC6295859 DOI: 10.1021/acsmedchemlett.8b00566] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Activation of Toll-like receptors (TLRs) located on immune cells leads to induction of immune responses that can be useful in vaccines for infectious diseases, cancer immunotherapy, and autoimmune diseases. Novel TLR signaling pathway modulators can further enhance the efficacy of TLR ligands.
Collapse
Affiliation(s)
- Nikunj
M. Shukla
- Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| | - Michael Chan
- Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| | - Tomoko Hayashi
- Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| | - Dennis A. Carson
- Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| | - Howard B. Cottam
- Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| |
Collapse
|
15
|
Sato-Kaneko F, Wang X, Yao S, Hosoya T, Lao FS, Messer K, Pu M, Shukla NM, Cottam HB, Chan M, Carson DA, Corr M, Hayashi T. Discovery of a Novel Microtubule Targeting Agent as an Adjuvant for Cancer Immunotherapy. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8091283. [PMID: 30406141 PMCID: PMC6199861 DOI: 10.1155/2018/8091283] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/17/2018] [Indexed: 12/26/2022]
Abstract
For an activating immunotherapy such as adjuvants, a compound that can prolong immune stimulation may enhance efficacy. We leveraged data from two prior high throughput screens with NF-κB and interferon reporter cell lines to identify 4H-chromene-3-carbonitriles as a class of compounds that prolonged activation in both screens. We repurchased 23 of the most promising candidates. Out of these compounds we found #1 to be the most effective agent in stimulating the release of cytokines and chemokines from immune cells, including murine primary bone marrow derived dendritic cells. Mechanistically, #1 inhibited tubulin polymerization, and its effect on immune cell activation was abolished in cells mutated in the beta-tubulin gene (TUBB) encoding the site where colchicine binds. Treatment with #1 resulted in mitochondrial depolarization followed by mitogen-activated protein kinase activation. Because tubulin polymerization modulating agents have been used for chemotherapy to treat malignancy and #1 activated cytokine responses, we hypothesized that #1 could be effective for cancer immunotherapy. Intratumoral injection of #1 delayed tumor growth in a murine syngeneic model of head and neck cancer. When combined with PD-1 blockade, tumor growth slowed in the injected tumor nodule and there was an abscopal effect in an uninjected nodule on the contralateral flank, suggesting central antitumor immune activation. Thus, we identified a new class of tubulin depolymerizing agent that acts as both an innate and an adaptive immune activating agent and that limits solid tumor growth when used concurrently with a checkpoint inhibitor.
Collapse
Affiliation(s)
- Fumi Sato-Kaneko
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Xiaodong Wang
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Nanhai Ave 3688, Shenzhen, Guangdong 518060, China
| | - Shiyin Yao
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Tadashi Hosoya
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Fitzgerald S. Lao
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Karen Messer
- Division of Biostatistics, University of California San Diego, La Jolla 92093, USA
| | - Minya Pu
- Division of Biostatistics, University of California San Diego, La Jolla 92093, USA
| | - Nikunj M. Shukla
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Howard B. Cottam
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Michael Chan
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Dennis A. Carson
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Maripat Corr
- Department of Medicine, University of California San Diego, La Jolla 92093, USA
| | - Tomoko Hayashi
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| |
Collapse
|
16
|
Shukla NM, Arimoto KI, Yao S, Fan JB, Zhang Y, Sato-Kaneko F, Lao FS, Hosoya T, Messer K, Pu M, Cottam HB, Carson DA, Hayashi T, Zhang DE, Corr M. Identification of Compounds That Prolong Type I Interferon Signaling as Potential Vaccine Adjuvants. SLAS DISCOVERY 2018; 23:960-973. [PMID: 29751735 DOI: 10.1177/2472555218774308] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vaccines are reliant on adjuvants to enhance the immune stimulus, and type I interferons (IFNs) have been shown to be beneficial in augmenting this response. We were interested in identifying compounds that would sustain activation of an endogenous type I IFN response as a co-adjuvant. We began with generation of a human monocytic THP-1 cell line with an IFN-stimulated response element (ISRE)-β-lactamase reporter construct for high-throughput screening. Pilot studies were performed to optimize the parameters and conditions for this cell-based Förster resonance energy transfer (FRET) reporter assay for sustaining an IFN-α-induced ISRE activation signal. These conditions were confirmed in an initial pilot screen, followed by the main screen for evaluating prolongation of an IFN-α-induced ISRE activation signal at 16 h. Hit compounds were identified using a structure enrichment strategy based on chemoinformatic clustering and a naïve "Top X" approach. A select list of confirmed hits was then evaluated for toxicity and the ability to sustain IFN activity by gene and protein expression. Finally, for proof of concept, a panel of compounds was used to immunize mice as co-adjuvant with a model antigen and an IFN-inducing Toll-like receptor 4 agonist, lipopolysaccharide, as an adjuvant. Selected compounds significantly augmented antigen-specific immunoglobulin responses.
Collapse
Affiliation(s)
- Nikunj M Shukla
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Kei-Ichiro Arimoto
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Shiyin Yao
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Jun-Bao Fan
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Yue Zhang
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Fumi Sato-Kaneko
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Fitzgerald S Lao
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Tadashi Hosoya
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Karen Messer
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA.,2 Department of Family Medicine and Public Health, Division of Biostatistics and Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Minya Pu
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA.,2 Department of Family Medicine and Public Health, Division of Biostatistics and Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Howard B Cottam
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Dennis A Carson
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | | | - Dong-Er Zhang
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA.,3 Department of Pathology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Maripat Corr
- 4 Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
17
|
Chan M, Kakitsubata Y, Hayashi T, Ahmadi A, Yao S, Shukla NM, Oyama SY, Baba A, Nguyen B, Corr M, Suda Y, Carson DA, Cottam HB, Wakao M. Structure-Activity Relationship Studies of Pyrimido[5,4-b]indoles as Selective Toll-Like Receptor 4 Ligands. J Med Chem 2017; 60:9142-9161. [PMID: 29049886 DOI: 10.1021/acs.jmedchem.7b00797] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Previous high throughput screening studies led to the discovery of two novel, nonlipid-like chemotypes as Toll-like receptor 4 (TLR4) agonists. One of these chemotypes, the pyrimido[5,4-b]indoles, was explored for structure-activity relationship trends relative to production of TLR4 dependent cytokines/chemokines, resulting in a semioptimized lead (compound 1) that provided a starting point for further optimization studies. In this report, compounds belonging to three areas of structural modification were evaluated for biological activity using murine and human TLR4 reporter cells, primary murine bone marrow derived dendritic cells, and human peripheral blood mononuclear cells. The compounds bearing certain aryl groups at the C8 position, such as phenyl (36) and β-naphthyl (39), had potencies significantly greater than compound 1. Compound 36 displayed human TLR4 agonist activity at submicromolar concentrations. The computational analysis suggests that the improved potency of these C8-aryl derivatives may be the result of additional binding interactions at the interface of the TLR4/myeloid differentiation protein-2 (MD-2) complex.
Collapse
Affiliation(s)
- Michael Chan
- Moores Cancer Center, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| | - Yuhei Kakitsubata
- Department of Chemistry, Kagoshima University , 1-21-40 Kohrimoto, 890-0065, Kagoshima, Japan
| | - Tomoko Hayashi
- Moores Cancer Center, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| | - Alast Ahmadi
- Moores Cancer Center, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| | - Shiyin Yao
- Moores Cancer Center, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| | - Nikunj M Shukla
- Moores Cancer Center, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| | - Shin-Ya Oyama
- Department of Chemistry, Kagoshima University , 1-21-40 Kohrimoto, 890-0065, Kagoshima, Japan
| | - Akihito Baba
- Department of Chemistry, Kagoshima University , 1-21-40 Kohrimoto, 890-0065, Kagoshima, Japan
| | - Brandon Nguyen
- Moores Cancer Center, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| | - Maripat Corr
- Department of Medicine, University of California, San Diego 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| | - Yasuo Suda
- Department of Chemistry, Kagoshima University , 1-21-40 Kohrimoto, 890-0065, Kagoshima, Japan
| | - Dennis A Carson
- Moores Cancer Center, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92093-0695, United States.,Department of Medicine, University of California, San Diego 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| | - Howard B Cottam
- Moores Cancer Center, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92093-0695, United States.,Department of Medicine, University of California, San Diego 9500 Gilman Drive, La Jolla, California 92093-0695, United States
| | - Masahiro Wakao
- Moores Cancer Center, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92093-0695, United States.,Department of Chemistry, Kagoshima University , 1-21-40 Kohrimoto, 890-0065, Kagoshima, Japan
| |
Collapse
|