1
|
Walker DM, Lazarova TI, Riesinger SW, Poirier MC, Messier T, Cunniff B, Walker VE. WR1065 conjugated to thiol-PEG polymers as novel anticancer prodrugs: broad spectrum efficacy, synergism, and drug resistance reversal. Front Oncol 2023; 13:1212604. [PMID: 37576902 PMCID: PMC10419174 DOI: 10.3389/fonc.2023.1212604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/27/2023] [Indexed: 08/15/2023] Open
Abstract
The lack of anticancer agents that overcome innate/acquired drug resistance is the single biggest barrier to achieving a durable complete response to cancer therapy. To address this issue, a new drug family was developed for intracellular delivery of the bioactive aminothiol WR1065 by conjugating it to discrete thiol-PEG polymers: 4-star-PEG-S-S-WR1065 (4SP65) delivers four WR1065s/molecule and m-PEG6-S-S-WR1065 (1LP65) delivers one. Infrequently, WR1065 has exhibited anticancer effects when delivered via the FDA-approved cytoprotectant amifostine, which provides one WR1065/molecule extracellularly. The relative anticancer effectiveness of 4SP65, 1LP65, and amifostine was evaluated in a panel of 15 human cancer cell lines derived from seven tissues. Additional experiments assessed the capacity of 4SP65 co-treatments to potentiate the anticancer effectiveness and overcome drug resistance to cisplatin, a chemotherapeutic, or gefitinib, a tyrosine kinase inhibitor (TKI) targeting oncogenic EGFR mutations. The CyQUANT®-NF proliferation assay was used to assess cell viability after 48-h drug treatments, with the National Cancer Institute COMPARE methodology employed to characterize dose-response metrics. In normal human epithelial cells, 4SP65 or 1LP65 enhanced or inhibited cell growth but was not cytotoxic. In cancer cell lines, 4SP65 and 1LP65 induced dose-dependent cytostasis and cytolysis achieving 99% cell death at drug concentrations of 11.2 ± 1.2 µM and 126 ± 15.8 µM, respectively. Amifostine had limited cytostatic effects in 11/14 cancer cell lines and no cytolytic effects. Binary pairs of 4SP65 plus cisplatin or gefitinib increased the efficacy of each partner drug and surmounted resistance to cytolysis by cisplatin and gefitinib in relevant cancer cell lines. 4SP65 and 1LP65 were significantly more effective against TP53-mutant than TP53-wild-type cell lines, consistent with WR1065-mediated reactivation of mutant p53. Thus, 4SP65 and 1LP65 represent a unique prodrug family for innovative applications as broad-spectrum anticancer agents that target p53 and synergize with a chemotherapeutic and an EGFR-TKI to prevent or overcome drug resistance.
Collapse
Affiliation(s)
- Dale M. Walker
- The Burlington HC Research Group, Inc., Jericho, VT, United States
| | | | | | - Miriam C. Poirier
- Carcinogen–DNA Interactions Section, Laboratory of Cellular Carcinogenesis and Tumor Promotion, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Terri Messier
- Department of Pathology and Laboratory Medicine, Redox Biology and Pathology Program, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Brian Cunniff
- Department of Pathology and Laboratory Medicine, Redox Biology and Pathology Program, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Vernon E. Walker
- Department of Pathology and Laboratory Medicine, Redox Biology and Pathology Program, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| |
Collapse
|
2
|
Jassas RS, Naeem N, Sadiq A, Mehmood R, Alenazi NA, Al-Rooqi MM, Mughal EU, Alsantali RI, Ahmed SA. Current status of N-, O-, S-heterocycles as potential alkaline phosphatase inhibitors: a medicinal chemistry overview. RSC Adv 2023; 13:16413-16452. [PMID: 37274413 PMCID: PMC10233329 DOI: 10.1039/d3ra01888a] [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: 03/22/2023] [Accepted: 05/24/2023] [Indexed: 06/06/2023] Open
Abstract
Heterocycles are a class of compounds that have been found to be potent inhibitors of alkaline phosphatase (AP), an enzyme that plays a critical role in various physiological processes such as bone metabolism, cell growth and differentiation, and has been linked to several diseases such as cancer and osteoporosis. AP is a widely distributed enzyme, and its inhibition has been considered as a therapeutic strategy for the treatment of these diseases. Heterocyclic compounds have been found to inhibit AP by binding to the active site of the enzyme, thereby inhibiting its activity. Heterocyclic compounds such as imidazoles, pyrazoles, and pyridines have been found to be potent AP inhibitors and have been studied as potential therapeutics for the treatment of cancer, osteoporosis, and other diseases. However, the development of more potent and selective inhibitors that can be used as therapeutics for the treatment of various diseases is an ongoing area of research. Additionally, the study of the mechanism of action of heterocyclic AP inhibitors is an ongoing area of research, which could lead to the identification of new targets and new therapeutic strategies. The enzyme known as AP has various physiological functions and is present in multiple tissues and organs throughout the body. This article presents an overview of the different types of AP isoforms, their distribution, and physiological roles. It also discusses the structure and mechanism of AP, including the hydrolysis of phosphate groups. Furthermore, the importance of AP as a clinical marker for liver disease, bone disorders, and cancer is emphasized, as well as its use in the diagnosis of rare inherited disorders such as hypophosphatasia. The potential therapeutic applications of AP inhibitors for different diseases are also explored. The objective of this literature review is to examine the function of alkaline phosphatase in various physiological conditions and diseases, as well as analyze the structure-activity relationships of recently reported inhibitors. The present review summarizes the structure-activity relationship (SAR) of various heterocyclic compounds as AP inhibitors. The SAR studies of these compounds have revealed that the presence of a heterocyclic ring, particularly a pyridine, pyrimidine, or pyrazole ring, in the molecule is essential for inhibitory activity. Additionally, the substitution pattern and stereochemistry of the heterocyclic ring also play a crucial role in determining the potency of the inhibitor.
Collapse
Affiliation(s)
- Rabab S Jassas
- Department of Chemistry, Jamoum University College, Umm Al-Qura University Makkah 21955 Saudi Arabia
| | - Nafeesa Naeem
- Department of Chemistry, University of Gujrat Gujrat 50700 Pakistan
| | - Amina Sadiq
- Department of Chemistry, Govt. College Women University Sialkot 51300 Pakistan
| | - Rabia Mehmood
- Department of Chemistry, Govt. College Women University Sialkot 51300 Pakistan
| | - Noof A Alenazi
- Department of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University Al-kharj 11942 Saudi Arabia
| | - Munirah M Al-Rooqi
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University 21955 Makkah Saudi Arabia
| | | | - Reem I Alsantali
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University P.O. Box 11099 Taif 21944 Saudi Arabia
| | - Saleh A Ahmed
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University 21955 Makkah Saudi Arabia
| |
Collapse
|
3
|
Khan NA, Rashid F, Jadoon MSK, Jalil S, Khan ZA, Orfali R, Perveen S, Al-Taweel A, Iqbal J, Shahzad SA. Design, Synthesis, and Biological Evaluation of Novel Dihydropyridine and Pyridine Analogs as Potent Human Tissue Nonspecific Alkaline Phosphatase Inhibitors with Anticancer Activity: ROS and DNA Damage-Induced Apoptosis. Molecules 2022; 27:molecules27196235. [PMID: 36234774 PMCID: PMC9570995 DOI: 10.3390/molecules27196235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 12/05/2022] Open
Abstract
Small molecules with nitrogen-containing scaffolds have gained much attention due to their biological importance in the development of new anticancer agents. The present paper reports the synthesis of a library of new dihydropyridine and pyridine analogs with diverse pharmacophores. All compounds were tested against the human tissue nonspecific alkaline phosphatase (h-TNAP) enzyme. Most of the compounds showed excellent enzyme inhibition against h-TNAP, having IC50 values ranging from 0.49 ± 0.025 to 8.8 ± 0.53 µM, which is multi-fold higher than that of the standard inhibitor (levamisole = 22.65 ± 1.60 µM) of the h-TNAP enzyme. Furthermore, an MTT assay was carried out to evaluate cytotoxicity against the HeLa and MCF-7 cancer cell lines. Among the analogs, the most potent dihydropyridine-based compound 4d was selected to investigate pro-apoptotic behavior. The further analysis demonstrated that compound 4d played a significant role in inducing apoptosis through multiple mechanisms, including overproduction of reactive oxygen species, mitochondrial dysfunction, DNA damaging, and arrest of the cell cycle at the G1 phase by inhibiting CDK4/6. The apoptosis-inducing effect of compound 4d was studied through staining agents, microscopic, and flow cytometry techniques. Detailed structure–activity relationship (SAR) and molecular docking studies were carried out to identify the core structural features responsible for inhibiting the enzymatic activity of the h-TNAP enzyme. Moreover, fluorescence emission studies corroborated the binding interaction of compound 4d with DNA through a fluorescence titration experiment.
Collapse
Affiliation(s)
- Nazeer Ahmad Khan
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, University Road, Abbottabad 22060, Pakistan
| | - Faisal Rashid
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Muhammad Siraj Khan Jadoon
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Saquib Jalil
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Zulfiqar Ali Khan
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Raha Orfali
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Shagufta Perveen
- Department of Chemistry, School of Computer, Mathematical and Natural Sciences, Morgan State University, Baltimore, MD 21251, USA
| | - Areej Al-Taweel
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Sohail Anjum Shahzad
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, University Road, Abbottabad 22060, Pakistan
- Correspondence: or
| |
Collapse
|
4
|
Yekehfallah V, Pahlavanneshan S, Sayadmanesh A, Momtahan Z, Ma B, Basiri M. Generation and Functional Characterization of PLAP CAR-T Cells against Cervical Cancer Cells. Biomolecules 2022; 12:biom12091296. [PMID: 36139135 PMCID: PMC9496028 DOI: 10.3390/biom12091296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 09/05/2022] [Accepted: 09/11/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is one of the cancer treatment modalities that has recently shown promising results in treating hematopoietic malignancies. However, one of the obstacles that need to be addressed in solid tumors is the on-target and off-tumor cytotoxicity due to the lack of specific tumor antigens with low expression in healthy cells. Placental alkaline phosphatase (PLAP) is a shared placenta- and tumor-associated antigen (TAA) that is expressed in ovarian, cervical, colorectal, and prostate cancers and is negligible in normal cells. In this study, we constructed second-generation CAR T cells with a fully human scFv against PLAP antigen andthen evaluated the characteristics of PLAP CAR T cells in terms of tonic signaling and differentiation in comparison with ΔPLAP CAR T cells and CD19 CAR T cells. In addition, by co-culturing PLAP CAR T cells with HeLa and CaSki cells, we analyzed the tumor-killing functions and the secretion of anti-tumor molecules. Results showed that PLAP CAR T cells not only proliferated during co-culture with cancer cells but also eliminated them in vitro. We also observed increased secretion of IL-2, granzyme A, and IFN-γ by PLAP CAR T cells upon exposure to the target cells. In conclusion, PLAP CAR T cells are potential candidates for further investigation in cervical cancer and, potentially, other solid tumors.
Collapse
Affiliation(s)
- Vahid Yekehfallah
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665666311, Iran
| | - Saghar Pahlavanneshan
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran 1968917313, Iran
| | - Ali Sayadmanesh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665666311, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 5166653431, Iran
| | - Zahra Momtahan
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
| | - Bin Ma
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Clinical Stem Cell Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Correspondence: (B.M.); (M.B.); Tel.: +86-21-62933631 (B.M.); +98-21-40223417 (M.B.)
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665666311, Iran
- Correspondence: (B.M.); (M.B.); Tel.: +86-21-62933631 (B.M.); +98-21-40223417 (M.B.)
| |
Collapse
|
5
|
Basiri M, Pahlavanneshan S. Evaluation of Placental Alkaline Phosphatase Expression as A Potential Target of Solid Tumors Immunotherapy by Using Gene and Protein Expression Repositories. CELL JOURNAL 2021; 23:717-721. [PMID: 34939766 PMCID: PMC8665984 DOI: 10.22074/cellj.2021.7299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 06/24/2020] [Indexed: 12/17/2022]
Abstract
Placental alkaline phosphatase (PLAP) is a membrane enzyme mainly expressed in the placenta. PLAP is shown to
be expressed in ovarian cancer (OV), however, there is little known about its expression in other cancers. Using gene
and protein expression deposited data, we surveyed PLAP expression across malignant and normal human tissues to
explore the potential of PLAP as an immunotherapy target. We detected more than two-fold increased PLAP expression
in multiple solid tumors including ovarian cancer, testicular germ cell tumors (TGCT), and uterine corpus endometrial
carcinoma (UCEC) compared with matched normal tissues. We also showed association of PLAP expression with high
mortality pancreatic adenocarcinoma (PAAD). Altogether, our results suggest that PLAP can be a promising target for
immunotherapy of multiple cancers, especially OV, TGCT, and UCEC.
Collapse
Affiliation(s)
- Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Saghar Pahlavanneshan
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Murer P, Plüss L, Neri D. A novel human monoclonal antibody specific to the A33 glycoprotein recognizes colorectal cancer and inhibits metastasis. MAbs 2021; 12:1714371. [PMID: 31928310 PMCID: PMC6999842 DOI: 10.1080/19420862.2020.1714371] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Colorectal cancer represents the second most common cause of cancer-related death. The human A33 transmembrane glycoprotein is a validated tumor-associated antigen, expressed in 95% of primary and metastatic colorectal cancers. Using phage display technology, we generated a human monoclonal antibody (termed A2) specific to human A33 and we compared its epitope and performance to those of previously described clinical-stage anti-human A33 antibodies. All antibodies recognized a similar immunodominant epitope, located in the V-domain of A33, as revealed by SPOT analysis. The A2 antibody homogenously stained samples of poorly, moderately, and well differentiated colon adenocarcinomas. All antibodies also exhibited an intense staining of healthy human colon sections. The A2 antibody, reformatted in murine IgG2a format, preferentially localized to A33-transfected CT26 murine colon adenocarcinomas in immunocompetent mice with a homogenous distribution within the tumor mass, while other antibodies exhibited a patchy uptake in neoplastic lesions. A2 efficiently induced killing of A33-expressing cells through antibody-dependent cell-mediated cytotoxicity in vitro and was able to inhibit the growth of A33-positive murine CT26 and C51 lung metastases in vivo. Anti-A33 antibodies may thus represent useful vehicles for the selective delivery of bioactive payloads to colorectal cancer, or may be used in IgG format in a setting of minimal residual disease.
Collapse
Affiliation(s)
- Patrizia Murer
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Louis Plüss
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| |
Collapse
|
7
|
Madaj R, Pawlowska R, Chworos A. In silico exploration of binding of selected bisphosphonate derivatives to placental alkaline phosphatase via docking and molecular dynamics. J Mol Graph Model 2020; 103:107801. [PMID: 33296741 DOI: 10.1016/j.jmgm.2020.107801] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/06/2020] [Accepted: 11/06/2020] [Indexed: 01/06/2023]
Abstract
Bisphosphonates constitute a group of pyrophosphate analogues therapeutically active against bone diseases. Numerous studies confirm their anticancer and antimetastatic potential as well as ability to relieve pathological pain. Although this is a known class of compounds, many aspects of their action remain unexplained and their new interaction partners are still being discovered. Due to the structural similarity to pyrophosphate, their interaction with pyrophosphate-recognizing enzymes seems to be feasible. In current work, the placental alkaline phosphatase (PLAP) is considered as a potential target for these class of compounds. PLAP is one of the enzymes responsible for degradation of pyrophosphate with high clinical significance. An elevation of PLAP level are considered as a potential cancer marker. An in silico study of complexes formed between selected phosphate derivatives and PLAP was performed. It indicates that all tested compounds: alendronic acid, clodronic acid, etidronic acid, zoledronic acid, imidodiphosphoric acid, pyrophosphoric acid, medronic acid, chloromethylenediphosphonic acid and hypophosphoric acid form a complexes with PLAP, stabilized by hydrogen bonds, hydrophobic and van der Waals interactions. Zoledronic acid, drug used in prevention of bone complications during cancer treatment was found to have the lowest estimated energy of binding (-6.6 kcal/mol). In silico study yielded very low energy of binding also for hypophosphate, equal -6.4 kcal/mol, despite having no identified hydrogen bonds. Subsequent molecular dynamic simulations, followed by molecular mechanics generalized-born surface area with pairwise decomposition calculations confirmed the stability of protein-ligand complexes. The results indicate that selected phosphate derivatives may potentially interact with the enzyme, changing its function, what should be investigated during in vitro studies.
Collapse
Affiliation(s)
- Rafal Madaj
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363, Lodz, Poland.
| | - Roza Pawlowska
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363, Lodz, Poland
| | - Arkadiusz Chworos
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363, Lodz, Poland
| |
Collapse
|
8
|
Su Y, Zhang X, Bidlingmaier S, Behrens CR, Lee NK, Liu B. ALPPL2 Is a Highly Specific and Targetable Tumor Cell Surface Antigen. Cancer Res 2020; 80:4552-4564. [PMID: 32868383 PMCID: PMC7572689 DOI: 10.1158/0008-5472.can-20-1418] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/29/2020] [Accepted: 08/19/2020] [Indexed: 11/16/2022]
Abstract
Identification of tumor-specific cell surface antigens has proven challenging, as the vast majority of tumor-associated antigens are also expressed in normal tissues. In mesothelioma, we identified a highly specific tumor cell surface antigen that can be targeted for therapy development. Mesothelioma is caused by malignant transformation of the mesothelium, is incurable, and can be categorized into three histologic subtypes: epithelioid, biphasic, and sarcomatoid. To identity novel mesothelioma cell surface antigens with broad subtype coverage and high tissue specificity, we have previously selected phage antibody display libraries on live mesothelioma cells and tissues following counterselection on normal cells and identified a panel of human antibodies that bind all subtypes of mesothelioma, but not normal mesothelium. One of the antibodies, M25, showed high specificity against an antigen we identify here as ALPPL2. IHC on normal human tissues found that ALPPL2 is expressed only on placental trophoblasts, but not on any other normal tissues. This significant tissue specificity and broad tumor type coverage suggest that ALPPL2 could be an excellent cell surface target for therapeutic development against mesothelioma. To evaluate therapeutic potential of ALPPL2 targeting, an ALPPL2-targeted antibody-drug conjugate was developed and demonstrated potent and specific tumor killing in vitro and in vivo against both epithelioid and sarcomatoid mesothelioma. Thus, ALPPL2 belongs to a rare class of cell surface antigens classified as truly tumor specific and is well suited for therapy development against ALPPL2-expressing tumors. SIGNIFICANCE: These findings identify ALPP2 as a true tumor-specific cell surface antigen whose tissue specificity enables the development of novel therapies.
Collapse
Affiliation(s)
- Yang Su
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Xin Zhang
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Scott Bidlingmaier
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Christopher R Behrens
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Nam-Kyung Lee
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, San Francisco, California.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| |
Collapse
|
9
|
Schuster J, Cheng SB, Padbury J, Sharma S. Placental extracellular vesicles and pre-eclampsia. Am J Reprod Immunol 2020; 85:e13297. [PMID: 32619308 DOI: 10.1111/aji.13297] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/16/2020] [Accepted: 06/26/2020] [Indexed: 12/16/2022] Open
Abstract
Pre-eclampsia is a hypertensive disease of pregnancy characterized by new-onset hypertension, with either proteinuria and/or organ dysfunction. Pre-eclampsia is a leading cause of maternal morbidity and mortality; however, the underlying cellular and molecular mechanisms are not well understood. There is consensus that the underlying mechanism(s) resulting in pre-eclampsia is centered around abnormal placentation, inadequate spiral-artery remodeling, and deficiency in trophoblast invasion, resulting in impaired maternal blood flow to the placenta and a release of signals and/or inflammatory mediators into maternal circulation triggering the systemic manifestations of pre-eclampsia. ER stress, resulting in impaired autophagy and placental release of aggregated proteins, may also confer systemic stress to maternal organs in pre-eclampsia. Extracellular vesicles (EVs), lipid-bilayer enclosed structures containing macromolecules including proteins, miRNA, and other important nucleotides, have been suggested to play an important role in this maternal-fetal communication. Circulating EVs are present in greater quantity in the plasma of pre-eclampsia subjects compared to normal pregnancy, and the placental derived EVs have been shown to have altered protein and RNA cargo. In this review, we will focus on EVs and their role in pre-eclampsia, specifically their role in immune responses, inflammation, altered angiogenesis, and endothelial dysfunction.
Collapse
Affiliation(s)
- Jessica Schuster
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Providence, RI, USA
| | - Shi-Bin Cheng
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Providence, RI, USA
| | - James Padbury
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Providence, RI, USA
| | - Surendra Sharma
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Providence, RI, USA
| |
Collapse
|
10
|
Wang J, Xu C, Cheng Q, Zhao J, Wu S, Li W, Ma W, Liu C, Jiang X. RNA Sequencing Revealed Signals of Evolution From Gallbladder Stone to Gallbladder Carcinoma. Front Oncol 2020; 10:823. [PMID: 32547950 PMCID: PMC7272658 DOI: 10.3389/fonc.2020.00823] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/28/2020] [Indexed: 12/24/2022] Open
Abstract
Gallbladder stone is a major risk factor for gallbladder carcinoma (GBC), while there is still a controversy whether period of follow-up since newly diagnoses of asymptomatic gallstones increases the risk of GBC. In this study, 10 GBC patients and 30 patients with gallstones were admitted to our hospital. Patients with gallstones were divided into 3 groups according to the follow-up time, involving 10 patients with follow-up period of 1–3 years (GS3 group), 10 patients with follow-up period of 5–10 years (GS5 group), and 10 patients with follow-up period of more than 10 years (GS10 group). Tumor and para-tumor tissues of GBC patients, and gallbladder tissues of gallstone patients were collected. RNA sequencing was performed on the 50 samples. Besides, 1,704 differentially expressed genes (DEGs) were identified in tumors compared with para-tumor tissues of 10 GBC patients, which were enriched into some well-known cancer-related pathways, such as PI3K-Akt, mitogen-activated protein kinase (MAPK), Ras, and Wnt signaling pathways, and the most significant pathway was neuroactive ligand-receptor interaction. Patients with gallstones with periods of follow-up equal to 1–3 and > 10 years showed to have higher cancer risk than those with 5–10 years. ALPP and GPR87 are potential biomarkers for predicting cancer risk in patients with gallstones. The in vitro results revealed that GPR-87 can promote the proliferation, migration, and invasion of GBC cells. Herein, we explored the relationship between GBC patients and patients with gallstones with different periods of follow-up in transcriptome level.
Collapse
Affiliation(s)
- Jinghan Wang
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Chang Xu
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Qingbao Cheng
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Jiangman Zhao
- Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China.,Shanghai Zhangjiang Institute of Medical Innovation, Shanghai, China
| | - Shouxin Wu
- Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China.,Shanghai Zhangjiang Institute of Medical Innovation, Shanghai, China
| | - Wushuang Li
- Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China.,Shanghai Zhangjiang Institute of Medical Innovation, Shanghai, China
| | - Wencong Ma
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Chen Liu
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Xiaoqing Jiang
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical University, Shanghai, China
| |
Collapse
|
11
|
Zaher DM, El‐Gamal MI, Omar HA, Aljareh SN, Al‐Shamma SA, Ali AJ, Zaib S, Iqbal J. Recent advances with alkaline phosphatase isoenzymes and their inhibitors. Arch Pharm (Weinheim) 2020; 353:e2000011. [DOI: 10.1002/ardp.202000011] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/09/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Dana M. Zaher
- Sharjah Institute for Medical ResearchSharjah United Arab Emirates
| | - Mohammed I. El‐Gamal
- Sharjah Institute for Medical ResearchSharjah United Arab Emirates
- College of PharmacySharjah United Arab Emirates
- Department of Medicinal ChemistryFaculty of PharmacyMansoura Egypt
| | - Hany A. Omar
- Sharjah Institute for Medical ResearchSharjah United Arab Emirates
- College of PharmacySharjah United Arab Emirates
- Department of PharmacologyFaculty of PharmacyBeni‐Suef Egypt
| | | | | | - Aya J. Ali
- College of PharmacySharjah United Arab Emirates
| | - Sumera Zaib
- Centre for Advanced Drug ResearchCOMSATS University Islamabad Abbottabad Campus Abbottabad Pakistan
| | - Jamshed Iqbal
- Centre for Advanced Drug ResearchCOMSATS University Islamabad Abbottabad Campus Abbottabad Pakistan
| |
Collapse
|
12
|
Paul A, Sil J. Identification of Differentially Expressed Genes to Establish New Biomarker for Cancer Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:1970-1985. [PMID: 29994718 DOI: 10.1109/tcbb.2018.2837095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The goal of the human genome project is to integrate genetic information into different clinical therapies. To achieve this goal, different computational algorithms are devised for identifying the biomarker genes, cause of complex diseases. However, most of the methods developed so far using DNA microarray data lack in interpreting biological findings and are less accurate in disease prediction. In the paper, we propose two parameters risk_factor and confusion_factor to identify the biologically significant genes for cancer development. First, we evaluate risk_factor of each gene and the genes with nonzero risk_factor result misclassification of data, therefore removed. Next, we calculate confusion_factor of the remaining genes which determines confusion of a gene in prediction due to closeness of the samples in the cancer and normal classes. We apply nondominated sorting genetic algorithm (NSGA-II) to select the maximally uncorrelated differentially expressed genes in the cancer class with minimum confusion_factor. The proposed Gene Selection Explore (GSE) algorithm is compared to well established feature selection algorithms using 10 microarray data with respect to sensitivity, specificity, and accuracy. The identified genes appear in KEGG pathway and have several biological importance.
Collapse
|
13
|
Doello K. The role of trophoblastic epigenetic reprogrammation in benign tumor cells on malignant progression: A molecular hypothesis. Med Hypotheses 2018. [PMID: 29523298 DOI: 10.1016/j.mehy.2018.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cancer tissues and placental ones share many properties such as invasiveness, metastasis and local immunosuppressive effects. The goal of the present article is to hypothesize a theory about cancer origin that links placental and cancerous tissues at molecular level. This hypothesis explain that cancer origin could be due to low hypoxic conditions in the peripheral zones of benign tumors which might up-regulate the expression of IGF2, and, consequently, trophoblastic genes. In fact, many phenotypic characteristics and molecular markers are shared between these two cell types (cancerous and trophoblastics ones), providing evidences to support this hypothesis. As a consequence, it could be interesting to demonstrate whether cancer start with a cellular reprogrammation towards a trophoblastic fate in order to design new antitumoral strategies focused on this fact.
Collapse
Affiliation(s)
- Kevin Doello
- Medical Oncology Service, Virgen de las Nieves Hospital (Granada), Av. Fuerzas Armadas, sn, 18014 Granada, Spain.
| |
Collapse
|
14
|
Gao X, Zhang Y, Saum KU, Schöttker B, Breitling LP, Brenner H. Tobacco smoking and smoking-related DNA methylation are associated with the development of frailty among older adults. Epigenetics 2016; 12:149-156. [PMID: 28001461 DOI: 10.1080/15592294.2016.1271855] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Tobacco smoking is a preventable environmental factor that contributes to a wide spectrum of age-related health outcomes; however, its association with the development of frailty is not yet well established. We examined the associations of self-reported smoking indicators, serum cotinine levels and smoking-related DNA methylation biomarkers with a quantitative frailty index (FI) in 2 independent subsets of older adults (age 50-75) recruited in Saarland, Germany in 2000 - 2002 (discovery set: n = 978, validation set: n = 531). We obtained DNA methylation profiles in whole blood samples by Illumina HumanMethylation450 BeadChip and calculated the FI according to the method of Mitnitski and Rockwood. Mixed linear regression models were implemented to assess the associations between smoking indicators and the FI. After controlling for potential covariates, current smoking, cumulative smoking exposure (pack-years), and time after smoking cessation (years) were significantly associated with the FI (P-value < 0.05). In the discovery panel, 17 out of 151 previously identified smoking-related CpG sites were associated with the FI after correction for multiple testing (FDR < 0.05). Nine of them survived in the validation phase and were designated as frailty-associated loci. A smoking index (SI) based on the 9 loci manifested a monotonic association with the FI. In conclusion, this study suggested that epigenetic alterations could play a role in smoking-associated development of frailty. The identified CpG sites have the potential to be prognostic biomarkers of frailty and frailty-related health outcomes. Our findings and the underlying mechanisms should be followed up in further, preferably longitudinal studies.
Collapse
Affiliation(s)
- Xu Gao
- a Division of Clinical Epidemiology and Aging Research , German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Yan Zhang
- a Division of Clinical Epidemiology and Aging Research , German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Kai-Uwe Saum
- a Division of Clinical Epidemiology and Aging Research , German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Ben Schöttker
- a Division of Clinical Epidemiology and Aging Research , German Cancer Research Center (DKFZ) , Heidelberg , Germany.,b Network Aging Research , University of Heidelberg , Heidelberg , Germany
| | - Lutz Philipp Breitling
- a Division of Clinical Epidemiology and Aging Research , German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Hermann Brenner
- a Division of Clinical Epidemiology and Aging Research , German Cancer Research Center (DKFZ) , Heidelberg , Germany.,b Network Aging Research , University of Heidelberg , Heidelberg , Germany.,c Division of Preventive Oncology , German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) , Heidelberg , Germany.,d German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) , Heidelberg , Germany
| |
Collapse
|
15
|
Weber M, Bujak E, Putelli A, Villa A, Matasci M, Gualandi L, Hemmerle T, Wulhfard S, Neri D. A highly functional synthetic phage display library containing over 40 billion human antibody clones. PLoS One 2014; 9:e100000. [PMID: 24950200 PMCID: PMC4065035 DOI: 10.1371/journal.pone.0100000] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 05/20/2014] [Indexed: 12/31/2022] Open
Abstract
Several synthetic antibody phage display libraries have been created and used for the isolation of human monoclonal antibodies. The performance of antibody libraries, which is usually measured in terms of their ability to yield high-affinity binding specificities against target proteins of interest, depends both on technical aspects (such as library size and quality of cloning) and on design features (which influence the percentage of functional clones in the library and their ability to be used for practical applications). Here, we describe the design, construction and characterization of a combinatorial phage display library, comprising over 40 billion human antibody clones in single-chain fragment variable (scFv) format. The library was designed with the aim to obtain highly stable antibody clones, which can be affinity-purified on protein A supports, even when used in scFv format. The library was found to be highly functional, as >90% of randomly selected clones expressed the corresponding antibody. When selected against more than 15 antigens from various sources, the library always yielded specific and potent binders, at a higher frequency compared to previous antibody libraries. To demonstrate library performance in practical biomedical research projects, we isolated the human antibody G5, which reacts both against human and murine forms of the alternatively spliced BCD segment of tenascin-C, an extracellular matrix component frequently over-expressed in cancer and in chronic inflammation. The new library represents a useful source of binding specificities, both for academic research and for the development of antibody-based therapeutics.
Collapse
Affiliation(s)
- Marcel Weber
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
- Philochem AG, Otelfingen, Switzerland
| | - Emil Bujak
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
- Philochem AG, Otelfingen, Switzerland
| | - Alessia Putelli
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
- Philochem AG, Otelfingen, Switzerland
| | | | | | | | | | | | - Dario Neri
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
- * E-mail:
| |
Collapse
|