1
|
Zlotnikov ID, Kudryashova EV. Targeted Polymeric Micelles System, Designed to Carry a Combined Cargo of L-Asparaginase and Doxorubicin, Shows Vast Improvement in Cytotoxic Efficacy. Polymers (Basel) 2024; 16:2132. [PMID: 39125158 PMCID: PMC11314107 DOI: 10.3390/polym16152132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
L-asparaginases (ASP) and Doxorubicin (Dox) are both used in the treatment of leukemia, including in combination. We have attempted to investigate if their combination within the same targeted delivery vehicle can make such therapy more efficacious. We assembled a micellar system, where the inner hydrophobic core was loaded with Dox, while ASP would absorb at the surface due to electrostatic interactions. To make such absorption stronger, we conjugated the ASP with oligoamines, such as spermine, and the lipid components of the micelle-lipoic and oleic acids-with heparin. When loaded with Dox alone, the system yielded about a 10-fold improvement in cytotoxicity, as compared to free Dox. ASP alone showed about a 2.5-fold increase in cytotoxicity, so, assuming additivity of the effect, one could expect a 25-fold improvement when the two agents are applied in combination. But in reality, a combination of ASP + Dox loaded into the delivery system produced a synergy, with a whopping 50× improvement vs. free individual component. Pharmacokinetic studies have shown prolonged circulation of micellar formulations in the bloodstream as well as an increase in the effective concentration of Dox in micellar form and a reduction in Dox accumulation to the liver and heart (which reduces hepatotoxicity and cardiotoxicity). For the same reason, Dox's liposomal formulation has been in use in the treatment of multiple types of cancer, almost replacing the free drug. We believe that an opportunity to deliver a combination of two types of drugs to the same target cell may represent a further step towards improvement in the risk-benefit ratio in cancer treatment.
Collapse
Affiliation(s)
| | - Elena V. Kudryashova
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia;
| |
Collapse
|
2
|
Andrade KCR, Homem-de-Mello M, Motta JA, Borges MG, de Abreu JAC, de Souza PM, Pessoa A, Pappas GJ, de Oliveira Magalhães P. A Structural In Silico Analysis of the Immunogenicity of L-Asparaginase from Penicillium cerradense. Int J Mol Sci 2024; 25:4788. [PMID: 38732010 PMCID: PMC11084778 DOI: 10.3390/ijms25094788] [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: 03/26/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
L-asparaginase is an essential drug used to treat acute lymphoid leukemia (ALL), a cancer of high prevalence in children. Several adverse reactions associated with L-asparaginase have been observed, mainly caused by immunogenicity and allergenicity. Some strategies have been adopted, such as searching for new microorganisms that produce the enzyme and applying protein engineering. Therefore, this work aimed to elucidate the molecular structure and predict the immunogenic profile of L-asparaginase from Penicillium cerradense, recently revealed as a new fungus of the genus Penicillium and producer of the enzyme, as a motivation to search for alternatives to bacterial L-asparaginase. In the evolutionary relationship, L-asparaginase from P. cerradense closely matches Aspergillus species. Using in silico tools, we characterized the enzyme as a protein fragment of 378 amino acids (39 kDa), including a signal peptide containing 17 amino acids, and the isoelectric point at 5.13. The oligomeric state was predicted to be a homotetramer. Also, this L-asparaginase presented a similar immunogenicity response (T- and B-cell epitopes) compared to Escherichia coli and Dickeya chrysanthemi enzymes. These results suggest a potentially useful L-asparaginase, with insights that can drive strategies to improve enzyme production.
Collapse
Affiliation(s)
- Kellen Cruvinel Rodrigues Andrade
- Laboratory of Natural Products, Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (K.C.R.A.)
| | - Mauricio Homem-de-Mello
- inSiliTox, Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia 70910-900, Brazil
| | - Julia Almeida Motta
- inSiliTox, Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia 70910-900, Brazil
| | - Marina Guimarães Borges
- Laboratory of Natural Products, Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (K.C.R.A.)
| | - Joel Antônio Cordeiro de Abreu
- Laboratory of Natural Products, Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (K.C.R.A.)
| | - Paula Monteiro de Souza
- Laboratory of Natural Products, Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (K.C.R.A.)
| | - Adalberto Pessoa
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Georgios J. Pappas
- Department Cell Biology, Institute Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil
| | - Pérola de Oliveira Magalhães
- Laboratory of Natural Products, Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (K.C.R.A.)
| |
Collapse
|
3
|
Mahboobi M, Salmanian AH, Sedighian H, Bambai B. Molecular Modeling and Optimization of Type II E.coli l-Asparginase Activity by in silico Design and in vitro Site-directed Mutagenesis. Protein J 2023; 42:664-674. [PMID: 37634213 DOI: 10.1007/s10930-023-10149-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION L-asparaginase (also known as L-ASNase) is a crucial therapeutic enzyme that is widely used in treatment of ALL (acute lymphoblastic leukemia) as a chemotherapeutic drug. Besides, this enzyme is used in the food industry as a food processing reagent to reduce the content of acrylamide in addition to the clinical industry. The improvement of activity and kinetic parameters of the L-ASNase enzyme may lead to higher efficiency resulting in practical achievement. In order to achieve this goal, we chosen glycine residue in position 88 as a potential mutation with advantageous outcomes. METHOD In this study, firstly to find the appropriate mutation on glycine 88, various in silico analyses, such as MD simulation and molecular docking, were carried out. Then, the rational design was adopted as the best strategy for molecular modifications of the enzyme to improve its enzymatic properties. RESULT Our in silico findings show that the four mutations G88Q, G88L, G88K, and G88A may be able to increase L-ASNase's asparaginase activity. The catalytic efficiency of each enzyme (kcat/Km) is the most important feature for comparing the catalytic activity of the mutants with the wild type form. The laboratory experiments showed that the kcat/Km for the G88Q mutant is 36.32% higher than the Escherichia coli K12 ASNase II (wild type), which suggests that L-ASNase activity is improved at lower concentration of L-ASN. Kinetic characterization of the mutants L-ASNase activity confirmed the high turnover rate (kcat) with ASN as substrate relative to the wild type enzyme. CONCLUSION In silico analyses and laboratory experiments demonstrated that the G88Q mutation rather than other mutation (G88L, G88K, and G88A) could improve the kinetics of L-ASNase.
Collapse
Affiliation(s)
- Mahdieh Mahboobi
- Department of Systems Biotechnology, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Shahrake- Pajoohesh Blvd., 15th Km, Tehran-Karaj Highway, P.O. Box 14965-161, Tehran, Iran
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali-Hatef Salmanian
- Department of Agricultural Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Shahrake- Pajoohesh Blvd., 15th Km, Tehran-Karaj Highway, P.O. Box 14965-161, Tehran, Iran.
| | - Hamid Sedighian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Bijan Bambai
- Department of Systems Biotechnology, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Shahrake- Pajoohesh Blvd., 15th Km, Tehran-Karaj Highway, P.O. Box 14965-161, Tehran, Iran.
| |
Collapse
|
4
|
Miranda J, Lefin N, Beltran JF, Belén LH, Tsipa A, Farias JG, Zamorano M. Enzyme Engineering Strategies for the Bioenhancement of L-Asparaginase Used as a Biopharmaceutical. BioDrugs 2023; 37:793-811. [PMID: 37698749 DOI: 10.1007/s40259-023-00622-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2023] [Indexed: 09/13/2023]
Abstract
Over the past few years, there has been a surge in the industrial production of recombinant enzymes from microorganisms due to their catalytic characteristics being highly efficient, selective, and biocompatible. L-asparaginase (L-ASNase) is an enzyme belonging to the class of amidohydrolases that catalyzes the hydrolysis of L-asparagine into L-aspartic acid and ammonia. It has been widely investigated as a biologic agent for its antineoplastic properties in treating acute lymphoblastic leukemia. The demand for L-ASNase is mainly met by the production of recombinant type II L-ASNase from Escherichia coli and Erwinia chrysanthemi. However, the presence of immunogenic proteins in L-ASNase sourced from prokaryotes has been known to result in adverse reactions in patients undergoing treatment. As a result, efforts are being made to explore strategies that can help mitigate the immunogenicity of the drug. This review gives an overview of recent biotechnological breakthroughs in enzyme engineering techniques and technologies used to improve anti-leukemic L-ASNase, taking into account the pharmacological importance of L-ASNase.
Collapse
Affiliation(s)
- Javiera Miranda
- Chemical Engineering Department, Universidad de la Frontera, Francisco Salazar 1145, 4811230, Temuco, Región de la Araucanía, Chile
| | - Nicolás Lefin
- Chemical Engineering Department, Universidad de la Frontera, Francisco Salazar 1145, 4811230, Temuco, Región de la Araucanía, Chile
| | - Jorge F Beltran
- Chemical Engineering Department, Universidad de la Frontera, Francisco Salazar 1145, 4811230, Temuco, Región de la Araucanía, Chile
| | - Lisandra Herrera Belén
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Santiago, Chile
| | - Argyro Tsipa
- Department of Civil and Environmental Engineering, University of Cyprus, Nicosia, Cyprus
| | - Jorge G Farias
- Chemical Engineering Department, Universidad de la Frontera, Francisco Salazar 1145, 4811230, Temuco, Región de la Araucanía, Chile
| | - Mauricio Zamorano
- Chemical Engineering Department, Universidad de la Frontera, Francisco Salazar 1145, 4811230, Temuco, Región de la Araucanía, Chile.
| |
Collapse
|
5
|
Shishparenok AN, Gladilina YA, Zhdanov DD. Engineering and Expression Strategies for Optimization of L-Asparaginase Development and Production. Int J Mol Sci 2023; 24:15220. [PMID: 37894901 PMCID: PMC10607044 DOI: 10.3390/ijms242015220] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Genetic engineering for heterologous expression has advanced in recent years. Model systems such as Escherichia coli, Bacillus subtilis and Pichia pastoris are often used as host microorganisms for the enzymatic production of L-asparaginase, an enzyme widely used in the clinic for the treatment of leukemia and in bakeries for the reduction of acrylamide. Newly developed recombinant L-asparaginase (L-ASNase) may have a low affinity for asparagine, reduced catalytic activity, low stability, and increased glutaminase activity or immunogenicity. Some successful commercial preparations of L-ASNase are now available. Therefore, obtaining novel L-ASNases with improved properties suitable for food or clinical applications remains a challenge. The combination of rational design and/or directed evolution and heterologous expression has been used to create enzymes with desired characteristics. Computer design, combined with other methods, could make it possible to generate mutant libraries of novel L-ASNases without costly and time-consuming efforts. In this review, we summarize the strategies and approaches for obtaining and developing L-ASNase with improved properties.
Collapse
Affiliation(s)
- Anastasiya N. Shishparenok
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (A.N.S.); (Y.A.G.)
| | - Yulia A. Gladilina
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (A.N.S.); (Y.A.G.)
| | - Dmitry D. Zhdanov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (A.N.S.); (Y.A.G.)
- Department of Biochemistry, Peoples’ Friendship University of Russia named after Patrice Lumumba (RUDN University), Miklukho—Maklaya St. 6, 117198 Moscow, Russia
| |
Collapse
|
6
|
Hozoorbakhsh F, Ghiasian M, Ghandehari F, Emami-Karvani Z, Khademi Dehkordi M. An immunoinformatic approach employing molecular docking and molecular dynamics simulation for evaluation of l-asparaginase produced by Bacillus velezensis. J Biomol Struct Dyn 2023; 41:9057-9071. [PMID: 36377397 DOI: 10.1080/07391102.2022.2139765] [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: 08/20/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022]
Abstract
l-Asparaginase is one of the most important treatments for acute lymphoblastic leukemia. In this study, l-asparaginase-producing bacteria were isolated from the effluent and soil of the Isfahan slaughterhouse using M9 specific medium. Isolates were identified by 16SrRNA phylogenetic analysis. The immune characteristics were predicted. Molecular docking was performed between l-asparaginase and l-asparagine substrate using AutoDock tools 4.2 and AutoDock Vina. Molecular dynamics simulation studies were fulfilled using GROMACS. Five l-asparaginase-producing bacteria isolated that belonging to Stenotrophomonas maltophilia, Chryseobacterium sp. Chryseobacterium indologenes, Bacillus velezensis and Bacillus safensis. Predictions showed B. velezensis has better immune characteristics than B. safensis. The binding energies of the docked complex were calculated to be -4.34 and -4.9 kcal/mol. Molecular docking confirmed the interaction of l-asparaginase with its substrate. It was observed that the residues Thr36, Tyr50, Ala47, Thr116, Asp117, Met142, Thr193 and Thr192 were fundamental in protein-ligand complexation. Also, RMSD, RMSF, Rg, DSSP, SASA and MM-PBSA analysis showed that when l-asparaginase is bound to l-asparagine, it did not lose stability, secondary structure and compactness. Slaughterhouse soils and effluents are a potential source of l-asparaginase-producing bacteria that probably can probably produce l-asparaginase with more favorable immune properties than commercial enzymes.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Mozhgan Ghiasian
- Department of Microbiology, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Fereshte Ghandehari
- Department of Microbiology, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | | | | |
Collapse
|
7
|
Pedroso A, Herrera Belén L, Beltrán JF, Castillo RL, Pessoa A, Pedroso E, Farías JG. In Silico Design of a Chimeric Humanized L-asparaginase. Int J Mol Sci 2023; 24:ijms24087550. [PMID: 37108713 PMCID: PMC10144303 DOI: 10.3390/ijms24087550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/13/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer among children worldwide, characterized by an overproduction of undifferentiated lymphoblasts in the bone marrow. The treatment of choice for this disease is the enzyme L-asparaginase (ASNase) from bacterial sources. ASNase hydrolyzes circulating L-asparagine in plasma, leading to starvation of leukemic cells. The ASNase formulations of E. coli and E. chrysanthemi present notorious adverse effects, especially the immunogenicity they generate, which undermine both their effectiveness as drugs and patient safety. In this study, we developed a humanized chimeric enzyme from E. coli L-asparaginase which would reduce the immunological problems associated with current L-asparaginase therapy. For these, the immunogenic epitopes of E. coli L-asparaginase (PDB: 3ECA) were determined and replaced with those of the less immunogenic Homo sapiens asparaginase (PDB:4O0H). The structures were modeled using the Pymol software and the chimeric enzyme was modeled using the SWISS-MODEL service. A humanized chimeric enzyme with four subunits similar to the template structure was obtained, and the presence of asparaginase enzymatic activity was predicted by protein-ligand docking.
Collapse
Affiliation(s)
- Alejandro Pedroso
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Temuco 4811230, Chile
| | - Lisandra Herrera Belén
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Avenida Carlos Schorr 255, Talca 3460000, Chile
| | - Jorge F Beltrán
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Temuco 4811230, Chile
| | - Rodrigo L Castillo
- Department of Internal Medicine, East Division, Faculty of Medicine, University of Chile, Santiago 7500922, Chile
| | - Adalberto Pessoa
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Enrique Pedroso
- Department of Family Medicine, Faculty of Medicine, University of Medical Sciences Matanzas, Matanzas 42300, Cuba
| | - Jorge G Farías
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Temuco 4811230, Chile
| |
Collapse
|
8
|
Tosta Pérez M, Herrera Belén L, Letelier P, Calle Y, Pessoa A, Farías JG. L-Asparaginase as the gold standard in the treatment of acute lymphoblastic leukemia: a comprehensive review. Med Oncol 2023; 40:150. [PMID: 37060469 DOI: 10.1007/s12032-023-02014-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/28/2023] [Indexed: 04/16/2023]
Abstract
L-Asparaginase is an antileukemic drug long approved for clinical use to treat childhood acute lymphoblastic leukemia, the most common cancer in this population worldwide. However, the efficacy and its use as a drug have been subject to debate due to the variety of adverse effects that patients treated with it present, as well as the prompt elimination in plasma, the need for multiple administrations, and high rates of allergic reactions. For this reason, the search for new, less immunogenic variants has long been the subject of study. This review presents the main aspects of the L-asparaginase enzyme from a structural, pharmacological, and clinical point of view, from the perspective of its use in chemotherapy protocols in conjunction with other drugs in the different treatment phases.
Collapse
Affiliation(s)
- María Tosta Pérez
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Temuco, Chile
| | - Lisandra Herrera Belén
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Santiago de Chile, Chile
| | - Pablo Letelier
- Precision Health Research Laboratory, Departamento de Procesos Diagnósticos y Evaluación, Facultad de Ciencias de La Salud, Universidad Católica de Temuco, Temuco, Chile
| | - Yolanda Calle
- Department of Life Sciences, Whitelands College, University of Roehampton, London, UK
| | - Adalberto Pessoa
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Jorge G Farías
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Temuco, Chile.
| |
Collapse
|
9
|
Belén LH, Beltrán JF, Pessoa A, Castillo RL, de Oliveira Rangel-Yagui C, Farías JG. Helicobacter pyloril-asparaginase: a study of immunogenicity from an in silico approach. 3 Biotech 2022; 12:286. [PMID: 36276451 PMCID: PMC9489821 DOI: 10.1007/s13205-022-03359-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/12/2022] [Indexed: 11/27/2022] Open
Abstract
Helicobacter pylori has become the causal agent of multiple forms of gastric disease worldwide, including gastric cancer. The enzyme l-asparaginase (ASNase) has been studied as a virulence factor. In this work, we performed an in silico investigation to characterize the immunological profile of H. pylori ASNase (HpASNase) to ascertain the possible implication of HpASNase immunogenicity in the H. pylori virulence mechanism. We applied a workflow based on bioinformatics tools, which, by calculating the relative frequency of immunogenic T-cell and B-cell epitopes, allowed us to predict the immunogenicity and allergenicity of HpASNase in silico. We also visualized the epitopes by mapping them into the native structure of the enzyme. We report for the first time the T-cell and B-cell epitope composition that contributes to the immunogenicity of this HpASNase, as well as the regions that could generate a hypersensitivity response in humans. ASNase from H. pylori resulted in highly immunogenic and allergenic. The high immunogenicity of HpASNase could imply the pathogenic mechanisms of H. pylori. This knowledge could be important for the development of new drugs against H. pylori infections. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03359-0.
Collapse
Affiliation(s)
- Lisandra Herrera Belén
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Santiago, Chile
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Avda. Francisco Salazar 01145, P.O. Box: 54-D, Temuco, Chile
| | - Jorge F. Beltrán
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Avda. Francisco Salazar 01145, P.O. Box: 54-D, Temuco, Chile
| | - Adalberto Pessoa
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, Universidad de Sao Paulo, São Paulo, Brazil
| | - Rodrigo L. Castillo
- Department of Internal Medicine East, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Carlota de Oliveira Rangel-Yagui
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, Universidad de Sao Paulo, São Paulo, Brazil
| | - Jorge G. Farías
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Avda. Francisco Salazar 01145, P.O. Box: 54-D, Temuco, Chile
| |
Collapse
|
10
|
Dhankhar R, Kawatra A, Gupta V, Mohanty A, Gulati P. In silico and in vitro analysis of arginine deiminase from Pseudomonas furukawaii as a potential anticancer enzyme. 3 Biotech 2022; 12:220. [PMID: 35971334 PMCID: PMC9374873 DOI: 10.1007/s13205-022-03292-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 07/30/2022] [Indexed: 11/24/2022] Open
Abstract
Arginine deiminase (ADI), a promising anticancer enzyme from Mycoplasma hominis, is currently in phase III of clinical trials for the treatment of arginine auxotrophic tumors. However, it has been associated with several drawbacks in terms of low stability at human physiological conditions, high immunogenicity, hypersensitivity and systemic toxicity. In our previous work, Pseudomonas furukawaii 24 was identified as a potent producer of ADI with optimum activity under physiological conditions. In the present study, phylogenetic analysis of microbial ADIs indicated P. furukawaii ADI (PfADI) to be closely related to experimentally characterized ADIs of Pseudomonas sp. with proven anticancer activity. Immunoinformatics analysis was performed indicating lower immunogenicity of PfADI than MhADI (M. hominis ADI) both in terms of number of linear and conformational B-cell epitopes and T-cell epitope density. Overall antigenicity and allergenicity of PfADI was also lower as compared to MhADI, suggesting the applicability of PfADI as an alternative anticancer biotherapeutic. Hence, in vitro experiments were performed in which the ADI coding arcA gene of P. furukawaii was cloned and expressed in E. coli BL21. Recombinant ADI of P. furukawaii was purified, characterized and its anticancer activity was assessed. The enzyme was stable at human physiological conditions (pH 7 and 37 °C) with Km of 1.90 mM. PfADI was found to effectively inhibit the HepG2 cells with an IC50 value of 0.1950 IU/ml. Therefore, the current in silico and in vitro studies establish PfADI as a potential anticancer drug candidate with improved efficacy and low immunogenicity. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03292-2.
Collapse
Affiliation(s)
- Rakhi Dhankhar
- Medical Microbiology and Bioprocess Technology Laboratory, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana India
| | - Anubhuti Kawatra
- Medical Microbiology and Bioprocess Technology Laboratory, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana India
| | - Vatika Gupta
- Medical Microbiology and Bioprocess Technology Laboratory, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana India
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Aparajita Mohanty
- Bioinformatics Infrastructure Facility, Gargi College, University of Delhi, New Delhi, India
| | - Pooja Gulati
- Medical Microbiology and Bioprocess Technology Laboratory, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana India
| |
Collapse
|
11
|
Śliwa-Tytko P, Kaczmarska A, Lejman M, Zawitkowska J. Neurotoxicity Associated with Treatment of Acute Lymphoblastic Leukemia Chemotherapy and Immunotherapy. Int J Mol Sci 2022; 23:ijms23105515. [PMID: 35628334 PMCID: PMC9146746 DOI: 10.3390/ijms23105515] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy is a milestone in the treatment of poor-prognosis pediatric acute lymphoblastic leukemia (ALL) and is expected to improve treatment outcomes and reduce doses of conventional chemotherapy without compromising the effectiveness of the therapy. However, both chemotherapy and immunotherapy cause side effects, including neurological ones. Acute neurological complications occur in 3.6–11% of children treated for ALL. The most neurotoxical chemotherapeutics are L-asparaginase (L-ASP), methotrexate (MTX), vincristine (VCR), and nelarabine (Ara-G). Neurotoxicity associated with methotrexate (MTX-NT) occurs in 3–7% of children treated for ALL and is characterized by seizures, stroke-like symptoms, speech disturbances, and encephalopathy. Recent studies indicate that specific polymorphisms in genes related to neurogenesis may have a predisposition to MTX toxicity. One of the most common complications associated with CAR T-cell therapy is immune effector cell-associated neurotoxicity syndrome (ICANS). Mechanisms of neurotoxicity in CAR T-cell therapy are still unknown and may be due to disruption of the blood–brain barrier and the effects of elevated cytokine levels on the central nervous system (CNS). In this review, we present an analysis of the current knowledge on the mechanisms of neurotoxicity of standard chemotherapy and the targeted therapy in children with ALL.
Collapse
Affiliation(s)
- Patrycja Śliwa-Tytko
- Student’s Scientific Association at the Department of Pediatric Hematology, Oncology and Transplantation, Medical University of Lublin, A. Racławickie 1, 20-059 Lublin, Poland;
| | - Agnieszka Kaczmarska
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Racławickie 1, 20-059 Lublin, Poland;
| | - Monika Lejman
- Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Racławickie 1, 20-059 Lublin, Poland; or
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, A. Racławickie 1, 20-059 Lublin, Poland
- Correspondence: or ; Tel.: +48-507-365-635
| |
Collapse
|
12
|
Masri M, Nur F, Widodo J, Jusuf E, Sahar W, Wahida N, Risnawati R, Nurbaya S, Asri TA, Fadly N. A novel L‐asparaginase from the symbiotic
Enterobacter aerogenes
isolated from
Eucheuma
sp. J FOOD PROCESS PRES 2022. [DOI: 10.1111/jfpp.16306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Mashuri Masri
- Department of Biology, Faculty of Science and Technology Alauddin State Islamic University Makassar Indonesia
| | - Fatmawati Nur
- Department of Biology, Faculty of Science and Technology Alauddin State Islamic University Makassar Indonesia
| | - Joko Widodo
- Laboratory of Technology Department STIKES Mega Rezky Makassar Indonesia
| | - Ekafadly Jusuf
- School of Management and Business (STIE) Amkop Makassar Indonesia
| | - Windy Sahar
- Department of Biology, Faculty of Science and Technology Alauddin State Islamic University Makassar Indonesia
| | - Nurul Wahida
- Department of Biology, Faculty of Science and Technology Alauddin State Islamic University Makassar Indonesia
| | - Risnawati Risnawati
- Department of Biology, Faculty of Science and Technology Alauddin State Islamic University Makassar Indonesia
| | - Siti Nurbaya
- Department of Biology, Faculty of Science and Technology Alauddin State Islamic University Makassar Indonesia
| | - Tuti Asriani Asri
- Department of Biology, Faculty of Science and Technology Alauddin State Islamic University Makassar Indonesia
| | - Nurul Fadly
- Department of Biology, Faculty of Science and Technology Alauddin State Islamic University Makassar Indonesia
| |
Collapse
|
13
|
Law ME, Davis BJ, Ghilardi AF, Yaaghubi E, Dulloo ZM, Wang M, Guryanova OA, Heldermon CD, Jahn SC, Castellano RK, Law BK. Repurposing Tranexamic Acid as an Anticancer Agent. Front Pharmacol 2022; 12:792600. [PMID: 35095503 PMCID: PMC8793890 DOI: 10.3389/fphar.2021.792600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/30/2021] [Indexed: 12/29/2022] Open
Abstract
Tranexamic Acid (TA) is a clinically used antifibrinolytic agent that acts as a Lys mimetic to block binding of Plasminogen with Plasminogen activators, preventing conversion of Plasminogen to its proteolytically activated form, Plasmin. Previous studies suggested that TA may exhibit anticancer activity by blockade of extracellular Plasmin formation. Plasmin-mediated cleavage of the CDCP1 protein may increase its oncogenic functions through several downstream pathways. Results presented herein demonstrate that TA blocks Plasmin-mediated excision of the extracellular domain of the oncoprotein CDCP1. In vitro studies indicate that TA reduces the viability of a broad array of human and murine cancer cell lines, and breast tumor growth studies demonstrate that TA reduces cancer growth in vivo. Based on the ability of TA to mimic Lys and Arg, we hypothesized that TA may perturb multiple processes that involve Lys/Arg-rich protein sequences, and that TA may alter intracellular signaling pathways in addition to blocking extracellular Plasmin production. Indeed, TA-mediated suppression of tumor cell viability is associated with multiple biochemical actions, including inhibition of protein synthesis, reduced activating phosphorylation of STAT3 and S6K1, decreased expression of the MYC oncoprotein, and suppression of Lys acetylation. Further, TA inhibited uptake of Lys and Arg by cancer cells. These findings suggest that TA or TA analogs may serve as lead compounds and inspire the production of new classes of anticancer agents that function by mimicking Lys and Arg.
Collapse
Affiliation(s)
- Mary E. Law
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
| | - Bradley J. Davis
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
| | - Amanda F. Ghilardi
- Department of Chemistry, University of Florida, Gainesville, FL, United States
| | - Elham Yaaghubi
- Department of Chemistry, University of Florida, Gainesville, FL, United States
| | - Zaafir M. Dulloo
- Department of Chemistry, University of Florida, Gainesville, FL, United States
| | - Mengxiong Wang
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
| | - Olga A. Guryanova
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
- UF Health Cancer Center, University of Florida, Gainesville, FL, United States
| | - Coy D. Heldermon
- UF Health Cancer Center, University of Florida, Gainesville, FL, United States
- Department of Medicine, University of Florida, Gainesville, FL, United States
| | - Stephan C. Jahn
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
| | - Ronald K. Castellano
- Department of Chemistry, University of Florida, Gainesville, FL, United States
- UF Health Cancer Center, University of Florida, Gainesville, FL, United States
| | - Brian K. Law
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
- UF Health Cancer Center, University of Florida, Gainesville, FL, United States
| |
Collapse
|
14
|
A Targeted Catalytic Nanobody (T-CAN) with Asparaginolytic Activity. Cancers (Basel) 2021; 13:cancers13225637. [PMID: 34830793 PMCID: PMC8616244 DOI: 10.3390/cancers13225637] [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: 10/21/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/25/2022] Open
Abstract
Simple Summary The therapy of Acute Lymphoblastic Leukemia (ALL) is based on Escherichia coli (E. coli) L-asparaginase, which is a very effective drug in most cases. However, its side effects sometimes prevent its usage or impose its interruption. The main issues derive from its bacterial origin, which elicits a strong immune response in the patient, and from its generalized action on all body compartments. In this work, we describe how we generated a fully active and miniaturized form of L-asparaginase starting from a camel single domain antibody, a class of antibodies known to have a very limited immunogenicity in humans. We also targeted it onto tumor cells by attaching it to an antibody fragment directed onto the CD19 B-cell surface receptor, expressed on ALL cells. We named this new molecule “Targeted Catalytic Nanobody” (T-CAN). The T-CAN is active and successfully binds to CD19 expressing cells in vitro. Thanks to its reduced immunogenic potential, it represents a new tool which deserves further development. Abstract E. coli L-asparaginase is an amidohydrolase (EC 3.5.1.1) which has been successfully used for the treatment of Acute Lymphoblastic Leukemia for over 50 years. Despite its efficacy, its side effects, and especially its intrinsic immunogenicity, hamper its usage in a significant subset of cases, thus limiting therapeutic options. Innovative solutions to improve on these drawbacks have been attempted, but none of them have been truly successful so far. In this work, we fully replaced the enzyme scaffold, generating an active, miniaturized form of L-asparaginase by protein engineering of a camel single domain antibody, a class of antibodies known to have a limited immunogenicity in humans. We then targeted it onto tumor cells by an antibody scFv fragment directed onto the CD19 B-cell surface receptor expressed on ALL cells. We named this new type of nanobody-based antibody-drug conjugate “Targeted Catalytic Nanobody” (T-CAN). The new molecule retains the catalytic activity and the binding capability of the original modules and successfully targets CD19 expressing cells in vitro. Thanks to its theoretically reduced immunogenic potential compared to the original molecule, the T-CAN can represent a novel approach to tackle current limitations in L-asparaginase usage.
Collapse
|
15
|
Liu Y, Yang W, Smith C, Cheng C, Karol SE, Larsen EC, Winick N, Carroll WL, Loh ML, Raetz EA, Hunger SP, Winter SS, Dunsmore KP, Devidas M, Yang JJ, Evans WE, Jeha S, Pui CH, Inaba H, Relling MV. Class II Human Leukocyte Antigen Variants Associate With Risk of Pegaspargase Hypersensitivity. Clin Pharmacol Ther 2021; 110:794-802. [PMID: 33768542 PMCID: PMC8790808 DOI: 10.1002/cpt.2241] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/13/2021] [Indexed: 10/20/2023]
Abstract
We conducted the first human leukocyte antigen (HLA) allele and genome-wide association study to identify loci associated with hypersensitivity reactions exclusively to the PEGylated preparation of asparaginase (pegaspargase) in racially diverse cohorts of pediatric leukemia patients: St Jude Children's Research Hospital's Total XVI (TXVI, n = 598) and Children's Oncology Group AALL0232 (n = 2,472) and AALL0434 (n = 1,189). Germline DNA was genotyped using arrays. Genetic variants not genotyped directly were imputed. HLA alleles were imputed using SNP2HLA or inferred using BWAkit. Analyses between genetic variants and hypersensitivity were performed in each cohort first using cohort-specific covariates and then combined using meta-analyses. Nongenetic risk factors included fewer intrathecal injections (P = 2.7 × 10-5 in TXVI) and male sex (P = 0.025 in AALL0232). HLA alleles DQB1*02:02, DRB1*07:01, and DQA1*02:01 had the strongest associations with pegaspargase hypersensitivity (P < 5.0 × 10-5 ) in patients with primarily European ancestry (EA), with the three alleles associating in a single haplotype. The top allele HLA-DQB1*02:02 was tagged by HLA-DQB1 rs1694129 in EAs (r2 = 0.96) and less so in non-EAs. All single nucleotide polymorphisms associated with pegaspargase hypersensitivity reaching genome-wide significance in EAs were in class II HLA loci, and were partially replicated in non-EAs, as is true for other HLA associations. The rs9958628 variant, in ARHGAP28 (previously linked to immune response in children) had the strongest genetic association (P = 8.9 × 10-9 ) in non-EAs. The HLA-DQB1*02:02-DRB1*07:01-DQA1*02:01 associated with hypersensitivity reactions to pegaspargase is the same haplotype associated with reactions to non-PEGylated asparaginase, even though the antigens differ between the two preparations.
Collapse
Affiliation(s)
- Yiwei Liu
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Wenjian Yang
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Colton Smith
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Cheng Cheng
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Seth E. Karol
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | | | - Naomi Winick
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
| | | | - Mignon L. Loh
- Department of Pediatrics, University of California School of Medicine, San Francisco, CA
| | | | - Stephen P. Hunger
- Department of Pediatrics, Children’s Hospital of Philadelphia and the Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA
| | - Stuart S. Winter
- Children’s Minnesota Cancer and Blood Disorders Program, Children’s Minnesota, Minneapolis, MN
| | | | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jun J. Yang
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - William E. Evans
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Sima Jeha
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Mary V. Relling
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
16
|
Low-cost agro-industrial sources as a substrate for the production of l-asparaginase using filamentous fungi. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2021. [DOI: 10.1016/j.bcab.2021.102037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
17
|
Circumventing the side effects of L-asparaginase. Biomed Pharmacother 2021; 139:111616. [PMID: 33932739 DOI: 10.1016/j.biopha.2021.111616] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022] Open
Abstract
L-asparaginase is an enzyme that catalyzes the degradation of asparagine and successfully used in the treatment of acute lymphoblastic leukemia. L-asparaginase toxicity is either related to hypersensitivity to the foreign protein or to a secondary L-glutaminase activity that causes inhibition of protein synthesis. PEGylated versions have been incorporated into the treatment protocols to reduce immunogenicity and an alternative L-asparaginase derived from Dickeya chrysanthemi is used in patients with anaphylactic reactions to the E. coli L-asparaginase. Alternative approaches commonly explore new sources of the enzyme as well as the use of protein engineering techniques to create less immunogenic, more stable variants with lower L-glutaminase activity. This article reviews the main strategies used to overcome L-asparaginase shortcomings and introduces recent tools that can be used to create therapeutic enzymes with improved features.
Collapse
|
18
|
Ameen F, Alshehri WA, Al-Enazi NM, Almansob A. L-Asparaginase activity analysis, ansZ gene identification and anticancer activity of a new Bacillus subtilis isolated from sponges of the Red Sea. Biosci Biotechnol Biochem 2020; 84:2576-2584. [DOI: 10.1080/09168451.2020.1807310] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Abstract
This study describes the isolation of various marine bacteriafrom sponges collected from the Red Sea (Saudi Arabia) andL-asparaginase (anti-cancer enzyme) production from bacterialisolates. The 16S rDNA based phylogenetic analysis revealed thatthe isolate WSA3 was a Bacillus subtilis. Its partial-length genesequence was submitted to GenBank under the accession numberMK072695. The new B. subtilis strain harbored the exact size(1128 bp) of the new L-asparaginase (ansZ) gene as confirmedby PCR and in gel visualization, which was submitted to the NCBIdatabase (accession number MN566442). The molecular weightof partially purified L-asparaginase was determined as 45 kDa bySDS-PAGE. In addition, the enzyme L-asparaginase did not showglutaminase activity which is very important from a medical pointof view. Moreover, 100 μg/mL of the partially purified B. subtilis Lasparaginaseshowed promising anti-cancer activities when testedagainst three cancer cell lines (HCT-116, MCF-7, and HepG2).
Collapse
Affiliation(s)
- Fuad Ameen
- Department of Botany & Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Wafa A Alshehri
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Nouf M Al-Enazi
- Department of Biology, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-kharj, Saudi Arabia
| | - Abobakr Almansob
- Department of Botany & Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
19
|
Costa-Silva T, Costa I, Biasoto H, Lima G, Silva C, Pessoa A, Monteiro G. Critical overview of the main features and techniques used for the evaluation of the clinical applicability of L-asparaginase as a biopharmaceutical to treat blood cancer. Blood Rev 2020; 43:100651. [DOI: 10.1016/j.blre.2020.100651] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 12/14/2019] [Accepted: 12/23/2019] [Indexed: 12/16/2022]
|
20
|
What makes a good new therapeutic l-asparaginase? World J Microbiol Biotechnol 2019; 35:152. [DOI: 10.1007/s11274-019-2731-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
|