1
|
Tseng YH, Lin HP, Lin SY, Chen BM, Vo TNN, Yang SH, Lin YC, Prijovic Z, Czosseck A, Leu YL, Roffler SR. Engineering stable and non-immunogenic immunoenzymes for cancer therapy via in situ generated prodrugs. J Control Release 2024; 369:179-198. [PMID: 38368947 DOI: 10.1016/j.jconrel.2024.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/15/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Engineering human enzymes for therapeutic applications is attractive but introducing new amino acids may adversely affect enzyme stability and immunogenicity. Here we used a mammalian membrane-tethered screening system (ECSTASY) to evolve human lysosomal beta-glucuronidase (hBG) to hydrolyze a glucuronide metabolite (SN-38G) of the anticancer drug irinotecan (CPT-11). Three human beta-glucuronidase variants (hBG3, hBG10 and hBG19) with 3, 10 and 19 amino acid substitutions were identified that display up to 40-fold enhanced enzymatic activity, higher stability than E. coli beta-glucuronidase in human serum, and similar pharmacokinetics in mice as wild-type hBG. The hBG variants were two to three orders of magnitude less immunogenic than E. coli beta-glucuronidase in hBG transgenic mice. Intravenous administration of an immunoenzyme (hcc49-hBG10) targeting a sialyl-Tn tumor-associated antigen to mice bearing human colon xenografts significantly enhanced the anticancer activity of CPT-11 as measured by tumor suppression and mouse survival. Our results suggest that genetically-modified human enzymes represent a good alternative to microbially-derived enzymes for therapeutic applications.
Collapse
Affiliation(s)
- Yi-Han Tseng
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Hsuan-Pei Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Sung-Yao Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Bing-Mae Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | | | - Shih-Hung Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Chen Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Zeljko Prijovic
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade 11001, Serbia
| | - Andreas Czosseck
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Yu-Lin Leu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
2
|
Burnouf PA, Roffler SR, Wu CC, Su YC. Glucuronides: From biological waste to bio-nanomedical applications. J Control Release 2022; 349:765-782. [PMID: 35907593 DOI: 10.1016/j.jconrel.2022.07.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 11/30/2022]
Abstract
Long considered as no more than biological waste meant to be eliminated in urine, glucuronides have recently contributed to tremendous developments in the biomedical field, particularly against cancer. While glucuronide prodrugs monotherapy and antibody-directed enzyme prodrug therapy have been around for some time, new facets have emerged that combine the unique properties of glucuronides notably in the fields of antibody-drug conjugates and nanomedicine. In both cases, glucuronides are utilized as a vector to improve pharmacokinetics and confer localized activation of potent drugs at tumor sites while also decreasing systemic toxicity. Here we will discuss some of the most promising strategies using glucuronides to promote successful anti-tumor therapeutic treatments.
Collapse
Affiliation(s)
- Pierre-Alain Burnouf
- International Center for Wound Repair and Regeneration, National Cheng-Kung University, Tainan, Taiwan.
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chia-Ching Wu
- International Center for Wound Repair and Regeneration, National Cheng-Kung University, Tainan, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Cheng Su
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
3
|
Walther R, Rautio J, Zelikin AN. Prodrugs in medicinal chemistry and enzyme prodrug therapies. Adv Drug Deliv Rev 2017; 118:65-77. [PMID: 28676386 DOI: 10.1016/j.addr.2017.06.013] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 06/27/2017] [Accepted: 06/29/2017] [Indexed: 12/21/2022]
Abstract
Prodrugs are cunning derivatives of therapeutic agents designed to improve the pharmacokinetics profile of the drug. Within a prodrug, pharmacological activity of the drug is masked and is recovered within the human body upon bioconversion of the prodrug, a process that is typically mediated by enzymes. This concept is highly successful and a significant fraction of marketed therapeutic formulations is based on prodrugs. An advanced subset of prodrugs can be engineered such as to achieve site-specific bioconversion of the prodrug - to comprise the highly advantageous "enzyme prodrug therapy", EPT. Design of prodrugs for EPT is similar to the prodrugs in general medicinal use in that the pharmacological activity of the drug is masked, but differs significantly in that site-specific bioconversion is a prime consideration, and the enzymes typically used for EPT are non-mammalian and/or with low systemic abundance in the human body. This review focuses on the design of prodrugs for EPT in terms of the choice of an enzyme and the corresponding prodrug for bioconversion. We also discuss the recent success of "self immolative linkers" which significantly empower and diversify the prodrug design, and present methodologies for the design of prodrugs with extended blood residence time. The review aims to be of specific interest for medicinal chemists, biomedical engineers, and pharmaceutical scientists.
Collapse
|
4
|
Guillen KP, Ruben EA, Virani N, Harrison RG. Annexin-directed β-glucuronidase for the targeted treatment of solid tumors. Protein Eng Des Sel 2017; 30:85-94. [PMID: 27986920 PMCID: PMC5241760 DOI: 10.1093/protein/gzw063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 10/10/2016] [Accepted: 11/17/2016] [Indexed: 01/13/2023] Open
Abstract
Enzyme prodrug therapy has the potential to remedy the lack of selectivity associated with the systemic administration of chemotherapy. However, most current systems are immunogenic and constrained to a monotherapeutic approach. We developed a new class of fusion proteins centered about the human enzyme β-glucuronidase (βG), capable of converting several innocuous prodrugs into chemotherapeutics. We targeted βG to phosphatidylserine on tumor cells, tumor vasculature and metastases via annexin A1/A5. Phosphatidylserine shows promise as a universal marker for solid tumors and allows for tumor type-independent targeting. To create fusion proteins, human annexin A1/A5 was genetically fused to the activity-enhancing 16a3 mutant of human βG, expressed in chemically defined, fed-batch suspension culture, and chromatographically purified. All fusion constructs achieved >95% purity with yields up to 740 μg/l. Fusion proteins displayed cancer selective cell-surface binding with cell line-dependent binding stability. One fusion protein in combination with the prodrug SN-38 glucuronide was as effective as the drug SN-38 on Panc-1 pancreatic cancer cells and HAAE-1 endothelial cells, and demonstrated efficacy against MCF-7 breast cancer cells. βG fusion proteins effectively enable localized combination therapy that can be tailored to each patient via prodrug selection, with promising clinical potential based on their near fully human design.
Collapse
Affiliation(s)
- Katrin P Guillen
- Biomedical Engineering Program and School of Chemical, Biological and Materials Engineering, University of Oklahoma, 100 E. Boyd St., Norman, OK 73019, USA
| | - Eliza A Ruben
- Protein Production Core, Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, USA
| | - Needa Virani
- Biomedical Engineering Program and School of Chemical, Biological and Materials Engineering, University of Oklahoma, 100 E. Boyd St., Norman, OK 73019, USA
| | - Roger G Harrison
- Biomedical Engineering Program and School of Chemical, Biological and Materials Engineering, University of Oklahoma, 100 E. Boyd St., Norman, OK 73019, USA
- Stephenson Cancer Center, Health Sciences Center, University of Oklahoma, 800 Northeast 10th St., Oklahoma City, OK 73104, USA
| |
Collapse
|
5
|
Sharma SK, Bagshawe KD. Translating antibody directed enzyme prodrug therapy (ADEPT) and prospects for combination. Expert Opin Biol Ther 2016; 17:1-13. [DOI: 10.1080/14712598.2017.1247802] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Surinder K. Sharma
- Research Department of Oncology, UCL Cancer Institute, University College London, London, UK
| | | |
Collapse
|
6
|
Prijovich ZM, Burnouf PA, Chou HC, Huang PT, Chen KC, Cheng TL, Leu YL, Roffler SR. Synthesis and Antitumor Properties of BQC-Glucuronide, a Camptothecin Prodrug for Selective Tumor Activation. Mol Pharm 2016; 13:1242-50. [DOI: 10.1021/acs.molpharmaceut.5b00771] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | - Pierre-Alain Burnouf
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
- Taiwan
International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | | | - Ping-Ting Huang
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Kai-Chuan Chen
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Tian-Lu Cheng
- Faculty
of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yu-Lin Leu
- Chia Nan University, Tainan 71710, Taiwan
| | - Steve R. Roffler
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
7
|
Hsieh YT, Chen KC, Cheng CM, Cheng TL, Tao MH, Roffler SR. Impediments to enhancement of CPT-11 anticancer activity by E. coli directed beta-glucuronidase therapy. PLoS One 2015; 10:e0118028. [PMID: 25688562 PMCID: PMC4331512 DOI: 10.1371/journal.pone.0118028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 01/05/2015] [Indexed: 12/17/2022] Open
Abstract
CPT-11 is a camptothecin analog used for the clinical treatment of colorectal adenocarcinoma. CPT-11 is converted into the therapeutic anti-cancer agent SN-38 by liver enzymes and can be further metabolized to a non-toxic glucuronide SN-38G, resulting in low SN-38 but high SN-38G concentrations in the circulation. We previously demonstrated that adenoviral expression of membrane-anchored beta-glucuronidase could promote conversion of SN-38G to SN-38 in tumors and increase the anticancer activity of CPT-11. Here, we identified impediments to effective tumor therapy with E. coli that were engineered to constitutively express highly active E. coli beta-glucuronidase intracellularly to enhance the anticancer activity of CPT-11. The engineered bacteria, E. coli (lux/βG), could hydrolyze SN-38G to SN-38, increased the sensitivity of cultured tumor cells to SN-38G by about 100 fold and selectively accumulated in tumors. However, E. coli (lux/βG) did not more effectively increase CPT-11 anticancer activity in human tumor xenografts as compared to non-engineered E. coli. SN-38G conversion to SN-38 by E. coli (lux/βG) appeared to be limited by slow uptake into bacteria as well as by segregation of E. coli in necrotic regions of tumors that may be relatively inaccessible to systemically-administered drug molecules. Studies using a fluorescent glucuronide probe showed that significantly greater glucuronide hydrolysis could be achieved in mice pretreated with E. coli (lux/βG) by direct intratumoral injection of the glucuronide probe or by intratumoral lysis of bacteria to release intracellular beta-glucuronidase. Our study suggests that the distribution of beta-glucuronidase, and possibly other therapeutic proteins, in the tumor microenvironment might be an important barrier for effective bacterial-based tumor therapy. Expression of secreted therapeutic proteins or induction of therapeutic protein release from bacteria might therefore be a promising strategy to enhance anti-tumor activity.
Collapse
Affiliation(s)
- Yuan-Ting Hsieh
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Kai-Chuan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chiu-Min Cheng
- Department of Aquaculture, National Kaohsiung Marine University, Kaohsiung, Taiwan
| | - Tian-Lu Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mi-Hua Tao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Steve R. Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
8
|
Matikonda SS, Orsi DL, Staudacher V, Jenkins IA, Fiedler F, Chen J, Gamble AB. Bioorthogonal prodrug activation driven by a strain-promoted 1,3-dipolar cycloaddition. Chem Sci 2014; 6:1212-1218. [PMID: 29560207 PMCID: PMC5811098 DOI: 10.1039/c4sc02574a] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 11/07/2014] [Indexed: 12/21/2022] Open
Abstract
Bioorthogonal prodrug activation controlled by the reaction of a trans-cyclooctene with an azide-functionalized prodrug is presented.
Due to the formation of hydrolysis-susceptible adducts, the 1,3-dipolar cycloaddition between an azide and strained trans-cyclooctene (TCO) has been disregarded in the field of bioorthogonal chemistry. We report a method which uses the instability of the adducts to our advantage in a prodrug activation strategy. The reaction of trans-cyclooctenol (TCO-OH) with a model prodrug resulted in a rapid 1,3-dipolar cycloaddition with second-order rates of 0.017 M–1 s–1 and 0.027 M–1 s–1 for the equatorial and axial isomers, respectively, resulting in release of the active compound. 1H NMR studies showed that activation proceeded via a triazoline and imine, both of which are rapidly hydrolyzed to release the model drug. Cytotoxicity of a doxorubicin prodrug was restored in vitro upon activation with TCO-OH, while with cis-cyclooctenol (CCO-OH) no activation was observed. The data also demonstrates the potential of this reaction in organic synthesis as a mild orthogonal protecting group strategy for amino and hydroxyl groups.
Collapse
Affiliation(s)
| | - Douglas L Orsi
- School of Pharmacy , University of Otago , Dunedin , 9054 , New Zealand .
| | - Verena Staudacher
- School of Pharmacy , University of Otago , Dunedin , 9054 , New Zealand .
| | - Imogen A Jenkins
- School of Pharmacy , University of Otago , Dunedin , 9054 , New Zealand .
| | - Franziska Fiedler
- School of Pharmacy , University of Otago , Dunedin , 9054 , New Zealand .
| | - Jiayi Chen
- School of Pharmacy , University of Otago , Dunedin , 9054 , New Zealand .
| | - Allan B Gamble
- School of Pharmacy , University of Otago , Dunedin , 9054 , New Zealand .
| |
Collapse
|
9
|
|
10
|
Tranoy-Opalinski I, Legigan T, Barat R, Clarhaut J, Thomas M, Renoux B, Papot S. β-Glucuronidase-responsive prodrugs for selective cancer chemotherapy: an update. Eur J Med Chem 2014; 74:302-13. [PMID: 24480360 DOI: 10.1016/j.ejmech.2013.12.045] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/22/2013] [Accepted: 12/23/2013] [Indexed: 02/07/2023]
Abstract
The design of novel antitumor agents allowing the destruction of malignant cells while sparing healthy tissues is one of the major challenges in medicinal chemistry. In this context, the use of non-toxic prodrugs programmed to be selectively activated by beta-glucuronidase present at high concentration in the microenvironment of most solid tumors has attracted considerable attention. This review summarizes the major progresses that have been realized in this field over the past ten years. This includes the new prodrugs that have been designed to target a wide variety of anticancer drugs, the prodrugs employed in the course of a combined therapy, the dendritic glucuronide prodrugs and the concept of β-glucuronidase-responsive albumin binding prodrugs.
Collapse
Affiliation(s)
- Isabelle Tranoy-Opalinski
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France
| | - Thibaut Legigan
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France
| | - Romain Barat
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France
| | - Jonathan Clarhaut
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France; INSERM CIC 0802, CHU de Poitiers, 2 rue de la Milétrie, 86021 Poitiers, France
| | - Mikaël Thomas
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France
| | - Brigitte Renoux
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France
| | - Sébastien Papot
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France.
| |
Collapse
|
11
|
Ma P, Kanizaj N, Chan SA, Ollis DL, McLeod MD. The Escherichia coli glucuronylsynthase promoted synthesis of steroid glucuronides: improved practicality and broader scope. Org Biomol Chem 2014; 12:6208-14. [DOI: 10.1039/c4ob00984c] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Steroid glucuronides can be quickly and conveniently prepared on the milligram scale using theE. coliglucuronylsynthase enzyme followed by purification with solid-phase extraction.
Collapse
Affiliation(s)
- Paul Ma
- Research School of Chemistry
- Australian National University
- Canberra, Australia
| | - Nicholas Kanizaj
- Research School of Chemistry
- Australian National University
- Canberra, Australia
| | - Shu-Ann Chan
- Research School of Chemistry
- Australian National University
- Canberra, Australia
| | - David L. Ollis
- Research School of Chemistry
- Australian National University
- Canberra, Australia
| | - Malcolm D. McLeod
- Research School of Chemistry
- Australian National University
- Canberra, Australia
| |
Collapse
|
12
|
Chen KC, Schmuck K, Tietze LF, Roffler SR. Selective cancer therapy by extracellular activation of a highly potent glycosidic duocarmycin analogue. Mol Pharm 2013; 10:1773-82. [PMID: 23448264 DOI: 10.1021/mp300581u] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Conventional cancer chemotherapy is limited by systemic toxicity and poor selectivity. Tumor-selective activation of glucuronide prodrugs by beta-glucuronidase in the tumor microenvironment in a monotherapeutic approach is one promising way to increase cancer selectivity. Here we examined the cellular requirement for enzymatic activation as well as the in vivo toxicity and antitumor activity of a glucuronide prodrug of a potent duocarmycin analogue that is active at low picomolar concentrations. Prodrug activation by intracellular and extracellular beta-glucuronidase was investigated by measuring prodrug 2 cytotoxicity against human cancer cell lines that displayed different endogenous levels of beta-glucuronidase, as well as against beta-glucuronidase-deficient fibroblasts and newly established beta-glucuronidase knockdown cancer lines. In all cases, glucuronide prodrug 2 was 1000-5000 times less cytotoxic than the parent duocarmycin analogue regardless of intracellular levels of beta-glucuronidase. By contrast, cancer cells that displayed tethered beta-glucuronidase on their plasma membrane were 80-fold more sensitive to glucuronide prodrug 2, demonstrating that prodrug activation depended primarily on extracellular rather than intracellular beta-glucuronidase activity. Glucuronide prodrug 2 (2.5 mg/kg) displayed greater antitumor activity and less systemic toxicity in vivo than the clinically used drug carboplatin (50 mg/kg) to mice bearing human lung cancer xenografts. Intratumoral injection of an adenoviral vector expressing membrane-tethered beta-glucuronidase dramatically enhanced the in vivo antitumor activity of prodrug 2. Our data provide evidence that increasing extracellular beta-glucuronidase activity in the tumor microenvironment can boost the therapeutic index of a highly potent glucuronide prodrug.
Collapse
Affiliation(s)
- Kai-Chuan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | | | | | |
Collapse
|
13
|
Stachulski AV, Meng X. Glucuronides from metabolites to medicines: a survey of the in vivo generation, chemical synthesis and properties of glucuronides. Nat Prod Rep 2013; 30:806-48. [DOI: 10.1039/c3np70003h] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
14
|
Chen CP, Hsieh YT, Prijovich ZM, Chuang HY, Chen KC, Lu WC, Tseng Q, Leu YL, Cheng TL, Roffler SR. ECSTASY, an adjustable membrane-tethered/soluble protein expression system for the directed evolution of mammalian proteins. Protein Eng Des Sel 2012; 25:367-75. [PMID: 22691701 DOI: 10.1093/protein/gzs033] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We describe an adjustable membrane-tethered/soluble protein screening methodology termed ECSTASY (enzyme cleavable surface tethered all-purpose screening system) which combines the power of high-throughput fluorescence-activated cell sorting of membrane-tethered proteins with the flexibility of soluble assays for isolation of improved mammalian recombinant proteins. In this approach, retroviral transduction is employed to stably tether a library of protein variants on the surface of mammalian cells via a glycosyl phosphatidylinositol anchor. High-throughput fluorescence-activated cell sorting is used to array cells expressing properly folded and/or active protein variants on their surface into microtiter culture plates. After culture to expand individual clones, treatment of cells with phosphatidylinositol-phospholipase C releases soluble protein variants for multiplex measurement of protein concentration, activity and/or function. We utilized ECSTASY to rapidly generate human β-glucuronidase variants for cancer therapy by antibody-directed enzyme prodrug therapy with up to 30-fold greater potency to catalyze the hydrolysis of the clinically relevant camptothecin anti-cancer prodrug as compared with wild-type human β-glucuronidase. A variety of recombinant proteins could be adjustably displayed on fibroblasts, suggesting that ECSTASY represents a general, simple and versatile methodology for high-throughput screening to accelerate sequence activity-based evolution of mammalian proteins.
Collapse
Affiliation(s)
- Cheng-Pao Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
The application of antibodies as therapeutic agents in the treatment of cancer now represents a significant proportion of the oncology drug arena. Despite this success, the ability to engineer and exploit antibodies in many different formats is ensuring that new avenues for their therapeutic application are constantly being examined. This review examines a selection of novel antibody-based therapeutic strategies that are currently in late preclinical and clinical evaluation.
Collapse
|
16
|
Hess M, Stritzker J, Härtl B, Sturm JB, Gentschev I, Szalay AA. Bacterial glucuronidase as general marker for oncolytic virotherapy or other biological therapies. J Transl Med 2011; 9:172. [PMID: 21989091 PMCID: PMC3207905 DOI: 10.1186/1479-5876-9-172] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 10/11/2011] [Indexed: 11/23/2022] Open
Abstract
Background Oncolytic viral tumor therapy is an emerging field in the fight against cancer with rising numbers of clinical trials and the first clinically approved product (Adenovirus for the treatment of Head and Neck Cancer in China) in this field. Yet, until recently no general (bio)marker or reporter gene was described that could be used to evaluate successful tumor colonization and/or transgene expression in other biological therapies. Methods Here, a bacterial glucuronidase (GusA) encoded by biological therapeutics (e.g. oncolytic viruses) was used as reporter system. Results Using fluorogenic probes that were specifically activated by glucuronidase we could show 1) preferential activation in tumors, 2) renal excretion of the activated fluorescent compounds and 3) reproducible detection of GusA in the serum of oncolytic vaccinia virus treated, tumor bearing mice in several tumor models. Time course studies revealed that reliable differentiation between tumor bearing and healthy mice can be done as early as 9 days post injection of the virus. Regarding the sensitivity of the newly developed assay system, we could show that a single infected tumor cell could be reliably detected in this assay. Conclusion GusA therefore has the potential to be used as a general marker in the preclinical and clinical evaluation of (novel) biological therapies as well as being useful for the detection of rare cells such as circulating tumor cells.
Collapse
Affiliation(s)
- Michael Hess
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | |
Collapse
|