1
|
Lathwal E, Kumar S, Sharma V, Sharma A, Choudhury T, Mistry T, Nasare VD. Design and Synthesis of Propargyloxy Functionalized Pyrazole-Based Aurones: Exploring Their Potent Anticancer Properties Against AGS and MCF-7 Cell Lines. Chem Biodivers 2024:e202402186. [PMID: 39641371 DOI: 10.1002/cbdv.202402186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/08/2024] [Accepted: 12/03/2024] [Indexed: 12/07/2024]
Abstract
FDA-approved numerous commercial and natural drugs used in cancer treatment feature either pyrazole or alkyne moieties. On the basis of this, we designed and synthesized 20 novel propargyloxy-substituted pyrazole-based aurones (10a-j and 11a-j) and evaluated for their anticancer potential against cancerous MCF-7 and human gastric adenocarcinoma (AGS) cell lines, as well as normal cell line human embryonic kidney 293 (HEK-293), through MTT assay. Among these tested compounds, five (10d-f, 11e, and 11f) displayed potent cytotoxic properties for AGS cancer cell line with IC50 values ranging from 19.7 to 28.5 µM, better than the reference drugs leucovorin (IC50 = 30.8 µM) and oxaliplatin (IC50 = 29.8 µM). Furthermore, compounds 10b, 10c, 11a, 11c, and 11d demonstrated a significant cytotoxic potential against the MCF-7 cancer cell line with a single-digit micromolar IC50 potency (4.8-8.5 µM) compared to the standard drug paclitaxel (IC50 = 19.7 µM). The cytotoxic studies of above selected potent active hybrid compounds, against HEK-293, normal cell line, further highlight the potential use of 10c molecule (IC50 = 4.8 µM against MCF-7 cells) as an anticancer agent for breast cancer with a selectivity index of 2.597. The cytotoxic results were further supported by the molecular docking studies.
Collapse
Affiliation(s)
- Ekta Lathwal
- Department of Chemistry, Kurukshetra University, Kurukshetra, Haryana, India
- Department of Chemistry, Pt. Chiranji Lal Sharma Government College, Karnal, Haryana, India
| | - Suresh Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra, Haryana, India
| | - Vikas Sharma
- Department of Chemistry, Kurukshetra University, Kurukshetra, Haryana, India
- Department of Chemistry, Pt. Chiranji Lal Sharma Government College, Karnal, Haryana, India
| | - Arpana Sharma
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Trisha Choudhury
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Tanuma Mistry
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Vilas D Nasare
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| |
Collapse
|
2
|
Borrego EA, Guerena CD, Schiaffino Bustamante AY, Gutierrez DA, Valenzuela CA, Betancourt AP, Varela-Ramirez A, Aguilera RJ. A Novel Pyrazole Exhibits Potent Anticancer Cytotoxicity via Apoptosis, Cell Cycle Arrest, and the Inhibition of Tubulin Polymerization in Triple-Negative Breast Cancer Cells. Cells 2024; 13:1225. [PMID: 39056806 PMCID: PMC11274517 DOI: 10.3390/cells13141225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
In this study, we screened a chemical library to find potent anticancer compounds that are less cytotoxic to non-cancerous cells. This study revealed that pyrazole PTA-1 is a potent anticancer compound. Additionally, we sought to elucidate its mechanism of action (MOA) in triple-negative breast cancer cells. Cytotoxicity was analyzed with the differential nuclear staining assay (DNS). Additional secondary assays were performed to determine the MOA of the compound. The potential MOA of PTA-1 was assessed using whole RNA sequencing, Connectivity Map (CMap) analysis, in silico docking, confocal microscopy, and biochemical assays. PTA-1 is cytotoxic at a low micromolar range in 17 human cancer cell lines, demonstrating less cytotoxicity to non-cancerous human cells, indicating a favorable selective cytotoxicity index (SCI) for the killing of cancer cells. PTA-1 induced phosphatidylserine externalization, caspase-3/7 activation, and DNA fragmentation in triple-negative breast MDA-MB-231 cells, indicating that it induces apoptosis. Additionally, PTA-1 arrests cells in the S and G2/M phases. Furthermore, gene expression analysis revealed that PTA-1 altered the expression of 730 genes at 24 h (198 upregulated and 532 downregulated). A comparison of these gene signatures with those within CMap indicated a profile similar to that of tubulin inhibitors. Subsequent studies revealed that PTA-1 disrupts microtubule organization and inhibits tubulin polymerization. Our results suggest that PTA-1 is a potent drug with cytotoxicity to various cancer cells, induces apoptosis and cell cycle arrest, and inhibits tubulin polymerization, indicating that PTA-1 is an attractive drug for future clinical cancer treatment.
Collapse
Affiliation(s)
- Edgar A. Borrego
- The Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, USA; (C.D.G.); (A.Y.S.B.); (D.A.G.); (C.A.V.); (A.P.B.); (A.V.-R.)
- Department of Biological Sciences, The University of Texas El Paso, El Paso, TX 79968, USA
| | - Cristina D. Guerena
- The Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, USA; (C.D.G.); (A.Y.S.B.); (D.A.G.); (C.A.V.); (A.P.B.); (A.V.-R.)
- Department of Biological Sciences, The University of Texas El Paso, El Paso, TX 79968, USA
| | - Austre Y. Schiaffino Bustamante
- The Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, USA; (C.D.G.); (A.Y.S.B.); (D.A.G.); (C.A.V.); (A.P.B.); (A.V.-R.)
- Department of Biological Sciences, The University of Texas El Paso, El Paso, TX 79968, USA
| | - Denisse A. Gutierrez
- The Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, USA; (C.D.G.); (A.Y.S.B.); (D.A.G.); (C.A.V.); (A.P.B.); (A.V.-R.)
- Department of Biological Sciences, The University of Texas El Paso, El Paso, TX 79968, USA
| | - Carlos A. Valenzuela
- The Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, USA; (C.D.G.); (A.Y.S.B.); (D.A.G.); (C.A.V.); (A.P.B.); (A.V.-R.)
- Department of Biological Sciences, The University of Texas El Paso, El Paso, TX 79968, USA
| | - Ana P. Betancourt
- The Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, USA; (C.D.G.); (A.Y.S.B.); (D.A.G.); (C.A.V.); (A.P.B.); (A.V.-R.)
- Department of Biological Sciences, The University of Texas El Paso, El Paso, TX 79968, USA
| | - Armando Varela-Ramirez
- The Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, USA; (C.D.G.); (A.Y.S.B.); (D.A.G.); (C.A.V.); (A.P.B.); (A.V.-R.)
- Department of Biological Sciences, The University of Texas El Paso, El Paso, TX 79968, USA
| | - Renato J. Aguilera
- The Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, USA; (C.D.G.); (A.Y.S.B.); (D.A.G.); (C.A.V.); (A.P.B.); (A.V.-R.)
- Department of Biological Sciences, The University of Texas El Paso, El Paso, TX 79968, USA
| |
Collapse
|
3
|
Jain PM, Gutierrez DA, Kumar S, Aguilera RJ, Karki SS. Synthesis of Novel Pyrazole-Oxindole Conjugates with Cytotoxicity in Human Cancer Cells via Apoptosis. Chem Biodivers 2023; 20:e202300843. [PMID: 37501576 PMCID: PMC10938640 DOI: 10.1002/cbdv.202300843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 07/29/2023]
Abstract
A novel series of pyrazole-oxindole conjugates were prepared and characterized as potential cytotoxic agents by FT-IR, NMR and HR-MS. The cytotoxic activity of these compounds was tested in the Jurkat acute T cell leukemia, CEM acute lymphoblastic leukemia, MCF10 A mammary epithelial and MDA-MB 231 triple negative breast cancer cell lines. Among the tested conjugates, 5-methyl-3-((3-(1-phenyl)-3-(p-tolyl)-1H-pyrazol-4-yl)methylene)indolin-2-one 6h emerged as the most cytotoxic with a CC50 of 4.36+/-0.2 μM against Jurkat cells. The mechanism of cell death induced by 6h was investigated through the Annexin V-FITC assay via flow cytometry. Reactive oxygen species (ROS) accumulation, mitochondrial health and the cell cycle progression were also evaluated in cells exposed to 6h. Results demonstrated that 6h induces apoptosis in a dose-response manner, without generating ROS and/or altering mitochondrial health. In addition, 6h disrupted the cell cycle distribution causing an increase in DNA fragmentation (Sub G0-G1), and an arrest in the G0-G1 phase. Taken together, the 6h compound revealed a strong potential as an antineoplastic agent evidenced by its cytotoxicity in leukemia cells, the activation of apoptosis and restriction of the cell cycle progression.
Collapse
Affiliation(s)
- Pravesh M. Jain
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy (A Constituent Unit of KLE Academy of Higher Education & Research-Belagavi), Rajajinagar, Bengaluru, Karnataka, INDIA-560010
| | - Denisse A. Gutierrez
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA
| | - Sujeet Kumar
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy (A Constituent Unit of KLE Academy of Higher Education & Research-Belagavi), Rajajinagar, Bengaluru, Karnataka, INDIA-560010
| | - Renato J. Aguilera
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA
| | - Subhas S. Karki
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy (A Constituent Unit of KLE Academy of Higher Education & Research-Belagavi), Rajajinagar, Bengaluru, Karnataka, INDIA-560010
| |
Collapse
|
4
|
Salem ME, Mahrous EM, Ragab EA, Nafie MS, Dawood KM. Synthesis of novel mono- and bis-pyrazolylthiazole derivatives as anti-liver cancer agents through EGFR/HER2 target inhibition. BMC Chem 2023; 17:51. [PMID: 37291635 DOI: 10.1186/s13065-023-00921-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 02/23/2023] [Indexed: 06/10/2023] Open
Abstract
3-Bromoacetyl-4-(2-naphthoyl)-1-phenyl-1H-pyrazole (6) was synthesized from 2-acetylnaphthalene and was used as a new key building block for constructing the title targets. Thus, the reaction of 6 with the thiosemicarbazones 7a-d and 9-11 afforded the corresponding simple naphthoyl-(3-pyrazolyl)thiazole hybrids 8a-d and 12 ~ 14. The symmetric bis-(2-naphthoyl-pyrazol-3-yl)thiazol-2-yl)hydrazono)methyl)phenoxy)alkanes 18a-c and 21a-c were similarly synthesized from reaction of 6 with the appropriate bis-thiosemicarbazones 17a-c and 19a-c, respectively. The synthesized two series of simple and symmetrical bis-molecular hybrid merging naphthalene, thiazole, and pyrazole were evaluated for their cytotoxicity. Compounds 18b,c and 21a showed the most potent cytotoxicity (IC50 = 0.97-3.57 µM) compared to Lapatinib (IC50 = 7.45 µM). Additionally, they were safe (non-cytotoxic) against the THLE2 cells with higher IC50 values. Compounds 18c exhibited promising EGFR and HER-2 inhibitory activities with IC50 = 4.98 and 9.85 nM, respectively, compared to Lapatinib (IC50 = 6.1 and 17.2 nM). Apoptosis investigation revealed that 18c significantly activated apoptotic cell death in HepG2 cells, increasing the death rate by 63.6-fold and arresting cell proliferation at the S-phase. Compound 18c upregulated P53 by 8.6-fold, Bax by 8.9-fold, caspase-3,8,9 by 9, 2.3, and 7.6-fold, while it inhibited the Bcl-2 expression by 0.34-fold. Thereby, compound 18c exhibited promising cytotoxicity against EGFR/HER2 inhibition against liver cancer.
Collapse
Affiliation(s)
- Mostafa E Salem
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Esraa M Mahrous
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Eman A Ragab
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Mohamed S Nafie
- Department of Chemistry (Biochemistry program), Faculty of Science, Suez Canal University, Ismailia, 41522, Egypt
| | - Kamal M Dawood
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt.
| |
Collapse
|
5
|
Wu J, Li J, He Y, Huang J, Zhao X, Pan B, Wang Y, Cheng L, Han J. DrugSim2DR: systematic prediction of drug functional similarities in the context of specific disease for drug repurposing. Gigascience 2022; 12:giad104. [PMID: 38116825 PMCID: PMC10729734 DOI: 10.1093/gigascience/giad104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/23/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Traditional approaches to drug development are costly and involve high risks. The drug repurposing approach can be a valuable alternative to traditional approaches and has therefore received considerable attention in recent years. FINDINGS Herein, we develop a previously undescribed computational approach, called DrugSim2DR, which uses a network diffusion algorithm to identify candidate anticancer drugs based on a drug functional similarity network. The innovation of the approach lies in the drug-drug functional similarity network constructed in a manner that implicitly links drugs through their common biological functions in the context of a specific disease state, as the similarity relationships based on general states (e.g., network proximity or Jaccard index of drug targets) ignore disease-specific molecular characteristics. The drug functional similarity network may provide a reference for prediction of drug combinations. We describe and validate the DrugSim2DR approach through analysis of data on breast cancer and lung cancer. DrugSim2DR identified some US Food and Drug Administration-approved anticancer drugs, as well as some candidate drugs validated by previous studies in the literature. Moreover, DrugSim2DR showed excellent predictive performance, as evidenced by receiver operating characteristic analysis and multiapproach comparisons in various cancer datasets. CONCLUSIONS DrugSim2DR could accurately assess drug-drug functional similarity within a specific disease context and may more effectively prioritize disease candidate drugs. To increase the usability of our approach, we have developed an R-based software package, DrugSim2DR, which is freely available on CRAN (https://CRAN.R-project.org/package=DrugSim2DR).
Collapse
Affiliation(s)
- Jiashuo Wu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Ji Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yalan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Junling Huang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Xilong Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Bingyue Pan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yahui Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Junwei Han
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
6
|
Aguilera RJ. A virus changed my life. Mol Biol Cell 2022; 33:ae2. [PMID: 36441842 PMCID: PMC9727801 DOI: 10.1091/mbc.e22-08-0357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The E. E. Just Award commemorates the great African-American cell biologist Dr. Ernest Everett Just, who was a successful pioneer in an era of systemic exclusion of minorities in science and academia. Receiving this award is not only an honor but a recognition of my long-standing commitment to helping Persons Excluded due to Ethnicity or Race (PEERS) to achieve success in biomedical careers. As a proud member of this group, I have devoted most of my career to training underrepresented undergraduate and graduate students to pursue scientific careers. My early work as a molecular immunologist focused on the search for enzymes involved in antigen-receptor gene recombination, as well as the characterization of nuclear factors involved in recombination and the transcriptional regulation of the murine recombination-activating genes. Over the past two decades, my research has focused on discovering and evaluating novel anticancer agents that can be used to treat various cancer types.
Collapse
Affiliation(s)
- Renato J. Aguilera
- Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519,*Address correspondence to: Renato J. Aguilera ()
| |
Collapse
|
7
|
Hess JD, Macias LH, Gutierrez DA, Moran-Santibanez K, Contreras L, Medina S, Villanueva PJ, Kirken RA, Varela-Ramirez A, Penichet ML, Aguilera RJ. Identification of a Unique Cytotoxic Thieno[2,3-c]Pyrazole Derivative with Potent and Selective Anticancer Effects In Vitro. BIOLOGY 2022; 11:biology11060930. [PMID: 35741451 PMCID: PMC9219615 DOI: 10.3390/biology11060930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/03/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022]
Abstract
In recent years, the thienopyrazole moiety has emerged as a pharmacologically active scaffold with antitumoral and kinase inhibitory activity. In this study, high-throughput screening of 2000 small molecules obtained from the ChemBridge DIVERset library revealed a unique thieno[2,3-c]pyrazole derivative (Tpz-1) with potent and selective cytotoxic effects on cancer cells. Compound Tpz-1 consistently induced cell death at low micromolar concentrations (0.19 μM to 2.99 μM) against a panel of 17 human cancer cell lines after 24 h, 48 h, or 72 h of exposure. Furthermore, an in vitro investigation of Tpz-1's mechanism of action revealed that Tpz-1 interfered with cell cycle progression, reduced phosphorylation of p38, CREB, Akt, and STAT3 kinases, induced hyperphosphorylation of Fgr, Hck, and ERK 1/2 kinases, and disrupted microtubules and mitotic spindle formation. These findings support the continued exploration of Tpz-1 and other thieno[2,3-c]pyrazole-based compounds as potential small-molecule anticancer agents.
Collapse
Affiliation(s)
- Jessica D. Hess
- Department of Biological Sciences and Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, The University of Texas at El Paso (UTEP), El Paso, TX 79902, USA; (J.D.H.); (L.H.M.); (D.A.G.); (K.M.-S.); (L.C.); (S.M.); (P.J.V.); (R.A.K.); (A.V.-R.)
| | - Luca H. Macias
- Department of Biological Sciences and Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, The University of Texas at El Paso (UTEP), El Paso, TX 79902, USA; (J.D.H.); (L.H.M.); (D.A.G.); (K.M.-S.); (L.C.); (S.M.); (P.J.V.); (R.A.K.); (A.V.-R.)
| | - Denisse A. Gutierrez
- Department of Biological Sciences and Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, The University of Texas at El Paso (UTEP), El Paso, TX 79902, USA; (J.D.H.); (L.H.M.); (D.A.G.); (K.M.-S.); (L.C.); (S.M.); (P.J.V.); (R.A.K.); (A.V.-R.)
| | - Karla Moran-Santibanez
- Department of Biological Sciences and Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, The University of Texas at El Paso (UTEP), El Paso, TX 79902, USA; (J.D.H.); (L.H.M.); (D.A.G.); (K.M.-S.); (L.C.); (S.M.); (P.J.V.); (R.A.K.); (A.V.-R.)
| | - Lisett Contreras
- Department of Biological Sciences and Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, The University of Texas at El Paso (UTEP), El Paso, TX 79902, USA; (J.D.H.); (L.H.M.); (D.A.G.); (K.M.-S.); (L.C.); (S.M.); (P.J.V.); (R.A.K.); (A.V.-R.)
| | - Stephanie Medina
- Department of Biological Sciences and Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, The University of Texas at El Paso (UTEP), El Paso, TX 79902, USA; (J.D.H.); (L.H.M.); (D.A.G.); (K.M.-S.); (L.C.); (S.M.); (P.J.V.); (R.A.K.); (A.V.-R.)
| | - Paulina J. Villanueva
- Department of Biological Sciences and Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, The University of Texas at El Paso (UTEP), El Paso, TX 79902, USA; (J.D.H.); (L.H.M.); (D.A.G.); (K.M.-S.); (L.C.); (S.M.); (P.J.V.); (R.A.K.); (A.V.-R.)
| | - Robert A. Kirken
- Department of Biological Sciences and Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, The University of Texas at El Paso (UTEP), El Paso, TX 79902, USA; (J.D.H.); (L.H.M.); (D.A.G.); (K.M.-S.); (L.C.); (S.M.); (P.J.V.); (R.A.K.); (A.V.-R.)
| | - Armando Varela-Ramirez
- Department of Biological Sciences and Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, The University of Texas at El Paso (UTEP), El Paso, TX 79902, USA; (J.D.H.); (L.H.M.); (D.A.G.); (K.M.-S.); (L.C.); (S.M.); (P.J.V.); (R.A.K.); (A.V.-R.)
| | - Manuel L. Penichet
- Division of Surgical Oncology, Department of Surgery and Department of Microbiology, Immunology and Molecular Genetics, The Molecular Biology Institute, AIDS Institute, Jonsson Comprehensive Cancer Center, The University of California, Los Angeles, CA 90095, USA;
| | - Renato J. Aguilera
- Department of Biological Sciences and Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, The University of Texas at El Paso (UTEP), El Paso, TX 79902, USA; (J.D.H.); (L.H.M.); (D.A.G.); (K.M.-S.); (L.C.); (S.M.); (P.J.V.); (R.A.K.); (A.V.-R.)
- Correspondence: ; Tel.: +1-915-747-6852
| |
Collapse
|