1
|
di Leandro L, Colasante M, Pitari G, Ippoliti R. Hosts and Heterologous Expression Strategies of Recombinant Toxins for Therapeutic Purposes. Toxins (Basel) 2023; 15:699. [PMID: 38133203 PMCID: PMC10748335 DOI: 10.3390/toxins15120699] [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: 11/15/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
The production of therapeutic recombinant toxins requires careful host cell selection. Bacteria, yeast, and mammalian cells are common choices, but no universal solution exists. Achieving the delicate balance in toxin production is crucial due to potential self-intoxication. Recombinant toxins from various sources find applications in antimicrobials, biotechnology, cancer drugs, and vaccines. "Toxin-based therapy" targets diseased cells using three strategies. Targeted cancer therapy, like antibody-toxin conjugates, fusion toxins, or "suicide gene therapy", can selectively eliminate cancer cells, leaving healthy cells unharmed. Notable toxins from various biological sources may be used as full-length toxins, as plant (saporin) or animal (melittin) toxins, or as isolated domains that are typical of bacterial toxins, including Pseudomonas Exotoxin A (PE) and diphtheria toxin (DT). This paper outlines toxin expression methods and system advantages and disadvantages, emphasizing host cell selection's critical role.
Collapse
Affiliation(s)
| | | | | | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (L.d.L.); (M.C.); (G.P.)
| |
Collapse
|
2
|
Bortolotti M, Biscotti F, Zanello A, Bolognesi A, Polito L. New Insights on Saporin Resistance to Chemical Derivatization with Heterobifunctional Reagents. Biomedicines 2023; 11:biomedicines11041214. [PMID: 37189832 DOI: 10.3390/biomedicines11041214] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Saporin is a type 1 ribosome-inactivating protein widely used as toxic payload in the construction of targeted toxins, chimeric molecules formed by a toxic portion linked to a carrier moiety. Among the most used carriers, there are large molecules (mainly antibodies) and small molecules (such as neurotransmitters, growth factors and peptides). Some saporin-containing targeted toxins have been used for the experimental treatment of several diseases, giving very promising results. In this context, one of the reasons for the successful use of saporin lies in its resistance to proteolytic enzymes and to conjugation procedures. In this paper, we evaluated the influence of derivatization on saporin using three heterobifunctional reagents, namely 2-iminothiolane (2-IT), N-succinimidyl 3-(2-pyridyldithio)propionate (SPDP) and 4-succinimidyloxycarbonyl-α-methyl-α-[2-pyridyldithio]toluene (SMPT). In order to obtain the highest number of inserted -SH groups with the lowest reduction of saporin biological activities, we assessed the residual ability of saporin to inhibit protein synthesis, to depurinate DNA and to induce cytotoxicity after derivatization. Our results demonstrate that saporin maintains an excellent resistance to derivatization processes, especially with SPDP, and permit us to define reaction conditions, in which saporin biological properties may not be altered. Therefore, these findings provide useful information for the construction of saporin-based targeted toxins, especially with small carriers.
Collapse
Affiliation(s)
- Massimo Bortolotti
- Department of Medical and Surgical Sciences-DIMEC, General Pathology Section, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Francesco Biscotti
- Department of Medical and Surgical Sciences-DIMEC, General Pathology Section, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Andrea Zanello
- Department of Medical and Surgical Sciences-DIMEC, General Pathology Section, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Andrea Bolognesi
- Department of Medical and Surgical Sciences-DIMEC, General Pathology Section, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Letizia Polito
- Department of Medical and Surgical Sciences-DIMEC, General Pathology Section, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
3
|
Plant-Derived Type I Ribosome Inactivating Protein-Based Targeted Toxins: A Review of the Clinical Experience. Toxins (Basel) 2022; 14:toxins14080563. [PMID: 36006226 PMCID: PMC9412999 DOI: 10.3390/toxins14080563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/04/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Targeted toxins (TT) for cancer treatment are a class of hybrid biologic comprised of a targeting domain coupled chemically or genetically to a proteinaceous toxin payload. The targeting domain of the TT recognises and binds to a defined target molecule on the cancer cell surface, thereby delivering the toxin that is then required to internalise to an appropriate intracellular compartment in order to kill the target cancer cell. Toxins from several different sources have been investigated over the years, and the two TTs that have so far been licensed for clinical use in humans; both utilise bacterial toxins. Relatively few clinical studies have, however, been undertaken with TTs that utilise single-chain type I ribosome inactivating proteins (RIPs). This paper reviews the clinical experience that has so far been obtained for a range of TTs based on five different type I RIPs and concludes that the majority studied in early phase trials show significant clinical activity that justifies further clinical investigation. A range of practical issues relating to the further clinical development of TT’s are also covered briefly together with some suggested solutions to outstanding problems.
Collapse
|
4
|
Kim S, Shukla RK, Kim E, Cressman SG, Yu H, Baek A, Choi H, Kim A, Sharma A, Wang Z, Huang CA, Reneau JC, Boyaka PN, Liyanage NPM, Kim S. Comparison of CD3e Antibody and CD3e-sZAP Immunotoxin Treatment in Mice Identifies sZAP as the Main Driver of Vascular Leakage. Biomedicines 2022; 10:1221. [PMID: 35740248 PMCID: PMC9220018 DOI: 10.3390/biomedicines10061221] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 02/03/2023] Open
Abstract
Anti-CD3-epsilon (CD3e) monoclonal antibodies (mAbs) and CD3e immunotoxins (ITs) are promising targeted therapy options for various T-cell disorders. Despite significant advances in mAb and IT engineering, vascular leakage syndrome (VLS) remains a major dose-limiting toxicity for ITs and has been poorly characterized for recent "engineered" mAbs. This study undertakes a direct comparison of non-mitogenic CD3e-mAb (145-2C11 with Fc-silentTM murine IgG1: S-CD3e-mAb) and a new murine-version CD3e-IT (saporin-streptavidin (sZAP) conjugated with S-CD3e-mAb: S-CD3e-IT) and identifies their distinct toxicity profiles in mice. As expected, the two agents showed different modes of action on T cells, with S-CD3e-mAb inducing nearly complete modulation of CD3e on the cell surface, while S-CD3e-IT depleted the cells. S-CD3e-IT significantly increased the infiltration of polymorphonuclear leukocytes (PMNs) into the tissue parenchyma of the spleen and lungs, a sign of increased vascular permeability. By contrast, S-CD3e-mAbs-treated mice showed no notable signs of vascular leakage. Treatment with control ITs (sZAP conjugated with Fc-silent isotype antibodies) induced significant vascular leakage without causing T-cell deaths. These results demonstrate that the toxin portion of S-CD3e-IT, not the CD3e-binding portion (S-CD3e-mAb), is the main driver of vascular leakage, thus clarifying the molecular target for improving safety profiles in CD3e-IT therapy.
Collapse
Affiliation(s)
- Shihyoung Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Rajni Kant Shukla
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Sophie G. Cressman
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Hannah Yu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Alice Baek
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Hyewon Choi
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Alan Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Amit Sharma
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
- Department of Microbial Immunity and Infection, The Ohio State University, Columbus, OH 43210, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Zhirui Wang
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Division of Plastic & Reconstructive Surgery, 12700 East 19th Avenue, Aurora, CO 80045, USA; (Z.W.); (C.A.H.)
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Division of Transplant Surgery, 12700 East 19th Avenue, Aurora, CO 80045, USA
| | - Christene A. Huang
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Division of Plastic & Reconstructive Surgery, 12700 East 19th Avenue, Aurora, CO 80045, USA; (Z.W.); (C.A.H.)
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Division of Transplant Surgery, 12700 East 19th Avenue, Aurora, CO 80045, USA
| | - John C. Reneau
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA;
| | - Prosper N. Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Namal P. M. Liyanage
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
- Department of Microbial Immunity and Infection, The Ohio State University, Columbus, OH 43210, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Sanggu Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
- Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
5
|
Yang J, Wang C, Luo L, Li Z, Xu B, Guo L, Xie J. Highly sensitive MALDI-MS measurement of active ricin: insight from more potential deoxynucleobase-hybrid oligonucleotide substrates. Analyst 2021; 146:2955-2964. [PMID: 33949380 DOI: 10.1039/d0an02205e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Herein, we report an improved MALDI-MS method for active ricin to contribute toward countermeasures against its real threat to the public. Compared with commonly used DNA or RNA substrates, the deoxynucleobase-hybrid oligonucleotide (RNA_dA, Rd) substrate containing functional Gd[combining low line]A[combining low line]GA loop was revealed as a substrate with more potential and used for the first time in ricin measurement via MALDI-MS. The Rd sequence greatly prompted ricin to exhibit its catalytic activity as rRNA N-glycosylase in ex vitro condition, which was supported by molecular docking simulation and enzymatic parameters depicted in MALDI-MS. Furthermore, we discovered that a highly pure matrix was the most crucial parameter for enhancing the sensitivity, which addressed the major obstacle encountered in the oligo(deoxy)nucleotide measurement, i.e., the interfering alkali metal ion-adducted signals in MALDI-MS. After the optimization of pH and enzymatic reaction buffer composition in this ex vitro condition, this method can provide a wide linearity of up to three orders of magnitude, i.e., 1-5000 ng mL-1, and a high sensitivity of 1 ng mL-1 without any enrichment. Denatured and active ricin could be distinctly differentiated, and the application to practical samples from one international exercise and a soft drink proved the feasibility of this new method. We believe this MALDI-MS method can contribute to the first response to ricin occurrence events in public safety and security, as well as pave a new way for a deep understanding of ricin and other type II ribosome inactivating proteins involved toxicology.
Collapse
Affiliation(s)
- Jiewei Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| | - Chenyu Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China. and School of Pharmacy, Minzu University, Beijing, 100081, China
| | - Li Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China. and School of Public Health, Hebei Medical University, Shijiazhuang, Hebei Province 050017, China
| | - Zhi Li
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| | - Bin Xu
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| | - Lei Guo
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| | - Jianwei Xie
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| |
Collapse
|
6
|
The Effect of Small Molecule Pharmacological Agents on the Triterpenoid Saponin Induced Endolysosomal Escape of Saporin and a Saporin-Based Immunotoxin in Target Human Lymphoma Cells. Biomedicines 2021; 9:biomedicines9030300. [PMID: 33804080 PMCID: PMC8000476 DOI: 10.3390/biomedicines9030300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 12/24/2022] Open
Abstract
Triterpenoid saponins augment the cytotoxicity of saporin based immunotoxins. It is postulated that this results from a saponin-mediated increase in the endolysosomal escape of the toxin to the cytosol, but this remains to be confirmed. To address this issue, we used a number of pharmacological inhibitors of endocytic processes as probes to investigate the role played by saponin in the endolysosomal escape of fluorescently labeled saporin and a saporin based immunotoxin targeted against CD38 on human lymphoma and leukemia cell lines. Endolysosomal escape of the toxin was measured by flow cytometric pulse shape analysis. These results were compared to the effects of the various inhibitors on the saponin-mediated augmentation of toxin and immunotoxin cytotoxicity. Inhibitors of clathrin-mediated endocytosis, micropinocytosis, and endosomal acidification abrogated the saponin-induced increase in the endolysosomal escape of the toxin into the cytosol, suggesting that these processes may be involved in the internalization of saponin to the same endolysosomal vesicle as the toxin. Alternatively, these processes may play a direct role in the mechanism by which saponin promotes toxin escape from the endolysosomal compartment to the cytosol. Correlation with the effects of these inhibitors on the augmentation of cytotoxicity provides additional evidence that endolysosomal escape is involved in driving augmentation.
Collapse
|
7
|
Arslan I, Akgul H, Kara M. Saporin, a Polynucleotide-Adenosine Nucleosidase, May Be an Efficacious Therapeutic Agent for SARS-CoV-2 Infection. SLAS DISCOVERY 2020; 26:330-335. [PMID: 33155515 PMCID: PMC8940857 DOI: 10.1177/2472555220970911] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Saporin, a type I ribosome-inactivating protein from soapwort plant, is a potent protein synthesis inhibitor. Catalytically, saporin is a characteristic N-glycosidase, and it depurinates a specific adenine residue from a universally conserved loop of the major ribosomal RNA (rRNA) of eukaryotic cells. It is well-known that saporin induces apoptosis through different pathways, including ribotoxic stress response, cell signal transduction, genomic DNA fragmentation and RNA abasic lyase (RAlyase) activity, and NAD+ depletion by poly-(ADP)-ribose polymerase hyperactivation. Saporin's high enzymatic activity, high stability, and resistance to conjugation procedures make it a well-suited tool for immunotherapy approaches.In the present study, we focus on saporin-based targeted toxins that may be efficacious therapeutic agents for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Our discussed points suggest that saporin may be a strategic molecule for therapeutic knockout treatments and a powerful candidate for novel drugs in the struggle against coronavirus 2019 (COVID-19).
Collapse
Affiliation(s)
- Idris Arslan
- Biomedical Engineeing, Bulent Ecevit University, Incivez, Zonguldak, Turkey
| | - Hafize Akgul
- Graduate School of Natural and Applied Sciences, Nanotechnology Engineeing, Bulent Ecevit University, Incivez, Zonguldak, Turkey
| | - Murat Kara
- Graduate School of Natural and Applied Sciences, Nanotechnology Engineeing, Bulent Ecevit University, Incivez, Zonguldak, Turkey
| |
Collapse
|
8
|
Wensley HJ, Johnston DA, Smith WS, Holmes SE, Flavell SU, Flavell DJ. A Flow Cytometric Method to Quantify the Endosomal Escape of a Protein Toxin to the Cytosol of Target Cells. Pharm Res 2019; 37:16. [PMID: 31873810 PMCID: PMC6928089 DOI: 10.1007/s11095-019-2725-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/18/2019] [Indexed: 02/08/2023]
Abstract
Purpose The aim of this work was to develop a quantitative, flow cytometric method for tracking the endolysosomal escape of a fluorescently labelled saporin toxin. Methods Flow cytometric measurements of fluorescent pulse width and height were used to track the endocytic uptake into Daudi cells of a fluorescently labelled saporin toxin and the saporin based immunotoxin, OKT10-SAP. Subsequently, measurement of changes in pulse width were used to investigate the effect of a triterpenoid saponin on the endolysosomal escape of internalised toxin into the cytosol. Live cell confocal microscopy was used to validate the flow cytometry data. Results Increased endolysosomal escape of saporin and OKT10-SAP was observed by confocal microscopy in cells treated with saponin. Fluorescent pulse width measurements were also able to detect and quantify escape more sensitively than confocal microscopy. Saponin induced endolysosomal escape could be abrogated by treatment with chloroquine, an inhibitor of endolysosomal acidification. Chloroquine abrogation of escape was also mirrored by a concomitant abrogation of cytotoxicity. Conclusions Poor endolysosomal escape is often a rate limiting step for the cytosolic delivery of protein toxins and other macromolecules. Pulse width analysis offers a simple method to semi-quantify the endolysosomal escape of this and similar molecules into the cytosol. Electronic supplementary material The online version of this article (10.1007/s11095-019-2725-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Harrison J Wensley
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, SO16 6YD, UK.,Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - David A Johnston
- Biomedical Imaging Unit, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Wendy S Smith
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Suzanne E Holmes
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Sopsamorn U Flavell
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, SO16 6YD, UK.,University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - David J Flavell
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, SO16 6YD, UK. .,University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, SO16 6YD, UK.
| |
Collapse
|
9
|
Dianthin and Its Potential in Targeted Tumor Therapies. Toxins (Basel) 2019; 11:toxins11100592. [PMID: 31614697 PMCID: PMC6832487 DOI: 10.3390/toxins11100592] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 10/03/2019] [Indexed: 12/26/2022] Open
Abstract
Dianthin enzymes belong to ribosome-inactivating proteins (RIPs) of type 1, i.e., they only consist of a catalytic domain and do not have a cell binding moiety. Dianthin-30 is very similar to saporin-S3 and saporin-S6, two RIPs often used to design targeted toxins for tumor therapy and already tested in some clinical trials. Nevertheless, dianthin enzymes also exhibit differences to saporin with regard to structure, efficacy, toxicity, immunogenicity and production by heterologous expression. Some of the distinctions might make dianthin more suitable for targeted tumor therapies than other RIPs. The present review provides an overview of the history of dianthin discovery and illuminates its structure, function and role in targeted toxins. It further discusses the option to increase the efficacy of dianthin by endosomal escape enhancers.
Collapse
|
10
|
Giansanti F, Flavell DJ, Angelucci F, Fabbrini MS, Ippoliti R. Strategies to Improve the Clinical Utility of Saporin-Based Targeted Toxins. Toxins (Basel) 2018; 10:toxins10020082. [PMID: 29438358 PMCID: PMC5848183 DOI: 10.3390/toxins10020082] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/29/2018] [Accepted: 02/11/2018] [Indexed: 02/06/2023] Open
Abstract
Plant Ribosome-inactivating proteins (RIPs) including the type I RIP Saporin have been used for the construction of Immunotoxins (ITxs) obtained via chemical conjugation of the toxic domain to whole antibodies or by generating genetic fusions to antibody fragments/targeting domains able to direct the chimeric toxin against a desired sub-population of cancer cells. The high enzymatic activity, stability and resistance to conjugation procedures and especially the possibility to express recombinant fusions in yeast, make Saporin a well-suited tool for anti-cancer therapy approaches. Previous clinical work on RIPs-based Immunotoxins (including Saporin) has shown that several critical issues must be taken into deeper consideration to fully exploit their therapeutic potential. This review focuses on possible combinatorial strategies (chemical and genetic) to augment Saporin-targeted toxin efficacy. Combinatorial approaches may facilitate RIP escape into the cytosolic compartment (where target ribosomes are), while genetic manipulations may minimize potential adverse effects such as vascular-leak syndrome or may identify T/B cell epitopes in order to decrease the immunogenicity following similar strategies as those used in the case of bacterial toxins such as Pseudomonas Exotoxin A or as for Type I RIP Bouganin. This review will further focus on strategies to improve recombinant production of Saporin-based chimeric toxins.
Collapse
Affiliation(s)
- Francesco Giansanti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, I-67100 L'Aquila, Italy.
| | - David J Flavell
- The Simon Flavell Leukaemia Research Laboratory (Leukaemia Busters), Southampton General Hospital, Southampton, SO16 8AT, UK.
| | - Francesco Angelucci
- Department of Life, Health and Environmental Sciences, University of L'Aquila, I-67100 L'Aquila, Italy.
| | | | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, I-67100 L'Aquila, Italy.
| |
Collapse
|
11
|
Bolognesi A, Bortolotti M, Battelli MG, Polito L. Hyperuricaemia, Xanthine Oxidoreductase and Ribosome-Inactivating Proteins from Plants: The Contributions of Fiorenzo Stirpe to Frontline Research. Molecules 2017; 22:molecules22020206. [PMID: 28134797 PMCID: PMC6155646 DOI: 10.3390/molecules22020206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 01/23/2017] [Indexed: 11/16/2022] Open
Abstract
The enzymes called ribosome-inactivating proteins (RIPs) that are able to depurinate nucleic acids and arrest vital cellular functions, including protein synthesis, are still a frontline research field, mostly because of their promising medical applications. The contributions of Stirpe to the development of these studies has been one of the most relevant. After a short biographical introduction, an overview is offered of the main results obtained by his investigations during last 55 years on his main research lines: hyperuricaemia, xanthine oxidoreductase and RIPs.
Collapse
Affiliation(s)
- Andrea Bolognesi
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy.
| | - Massimo Bortolotti
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy.
| | - Maria Giulia Battelli
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy.
| | - Letizia Polito
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy.
| |
Collapse
|
12
|
Bolognesi A, Bortolotti M, Maiello S, Battelli MG, Polito L. Ribosome-Inactivating Proteins from Plants: A Historical Overview. Molecules 2016; 21:molecules21121627. [PMID: 27898041 PMCID: PMC6273060 DOI: 10.3390/molecules21121627] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 11/22/2016] [Accepted: 11/23/2016] [Indexed: 12/12/2022] Open
Abstract
This review provides a historical overview of the research on plant ribosome-inactivating proteins (RIPs), starting from the first studies at the end of eighteenth century involving the purification of abrin and ricin, as well as the immunological experiments of Paul Erlich. Interest in these plant toxins was revived in 1970 by the observation of their anticancer activity, which has given rise to a large amount of research contributing to the development of various scientific fields. Biochemistry analyses succeeded in identifying the enzymatic activity of RIPs and allowed for a better understanding of the ribosomal machinery. Studies on RIP/cell interactions were able to detail the endocytosis and intracellular routing of ricin, thus increasing our knowledge of how cells handle exogenous proteins. The identification of new RIPs and the finding that most RIPs are single-chain polypeptides, together with their genetic sequencing, has aided in the development of new phylogenetic theories. Overall, the biological properties of these proteins, including their abortifacient, anticancer, antiviral and neurotoxic activities, suggest that RIPs could be utilized in agriculture and in many biomedical fields, including clinical drug development.
Collapse
Affiliation(s)
- Andrea Bolognesi
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy.
| | - Massimo Bortolotti
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy.
| | - Stefania Maiello
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy.
| | - Maria Giulia Battelli
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy.
| | - Letizia Polito
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy.
| |
Collapse
|
13
|
Polito L, Djemil A, Bortolotti M. Plant Toxin-Based Immunotoxins for Cancer Therapy: A Short Overview. Biomedicines 2016; 4:biomedicines4020012. [PMID: 28536379 PMCID: PMC5344252 DOI: 10.3390/biomedicines4020012] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 05/23/2016] [Accepted: 05/30/2016] [Indexed: 02/07/2023] Open
Abstract
Immunotoxins are chimeric proteins obtained by linking a toxin to either an intact antibody or an antibody fragment. Conjugation can be obtained by chemical or genetic engineering, where the latter yields recombinant conjugates. An essential requirement is that the target molecule recognized by the antibody is confined to the cell population to be deleted, or at least that it is not present on stem cells or other cell types essential for the organism’s survival. Hundreds of different studies have demonstrated the potential for applying immunotoxins to many models in pre-clinical studies and in clinical trials. Immunotoxins can be theoretically used to eliminate any unwanted cell responsible for a pathological condition. The best results have been obtained in cancer therapy, especially in hematological malignancies. Among plant toxins, the most frequently employed to generate immunotoxins are ribosome-inactivating proteins, the most common being ricin. This review summarizes the various approaches and results obtained in the last four decades by researchers in the field of plant toxin-based immunotoxins for cancer therapy.
Collapse
Affiliation(s)
- Letizia Polito
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy.
| | - Alice Djemil
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy.
| | - Massimo Bortolotti
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy.
| |
Collapse
|
14
|
Sánchez M, Scirè A, Tanfani F, Ausili A. The thermal unfolding of the ribosome-inactivating protein saporin-S6 characterized by infrared spectroscopy. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1357-64. [DOI: 10.1016/j.bbapap.2015.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 06/08/2015] [Accepted: 06/17/2015] [Indexed: 10/23/2022]
|
15
|
Gilabert-Oriol R, Weng A, Mallinckrodt BV, Melzig MF, Fuchs H, Thakur M. Immunotoxins constructed with ribosome-inactivating proteins and their enhancers: a lethal cocktail with tumor specific efficacy. Curr Pharm Des 2014; 20:6584-643. [PMID: 25341935 PMCID: PMC4296666 DOI: 10.2174/1381612820666140826153913] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 08/05/2014] [Indexed: 11/30/2022]
Abstract
The term ribosome-inactivating protein (RIP) is used to denominate proteins mostly of plant origin, which have N-glycosidase enzymatic activity leading to a complete destruction of the ribosomal function. The discovery of the RIPs was almost a century ago, but their usage has seen transition only in the last four decades. With the advent of antibody therapy, the RIPs have been a subject of extensive research especially in targeted tumor therapies, which is the primary focus of this review. In the present work we enumerate 250 RIPs, which have been identified so far. An attempt has been made to identify all the RIPs that have been used for the construction of immunotoxins, which are conjugates or fusion proteins of an antibody or ligand with a toxin. The data from 1960 onwards is reviewed in this paper and an extensive list of more than 450 immunotoxins is reported. The clinical reach of tumor-targeted toxins has been identified and detailed in the work as well. While there is a lot of potential that RIPs embrace for targeted tumor therapies, the success in preclinical and clinical evaluations has been limited mainly because of their inability to escape the endo/lysosomal degradation. Various strategies that can increase the efficacy and lower the required dose for targeted toxins have been compiled in this article. It is plausible that with the advancements in platform technologies or improved endosomal escape the usage of tumor targeted RIPs would see the daylight of clinical success.
Collapse
Affiliation(s)
| | | | | | | | | | - Mayank Thakur
- Institut fur Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Charite - Universitatsmedizin Berlin, Campus Virchow-Klinikum (Forum 4), Augustenburger Platz 1, D-13353 Berlin, Germany.
| |
Collapse
|
16
|
Saporin-S6: a useful tool in cancer therapy. Toxins (Basel) 2013; 5:1698-722. [PMID: 24105401 PMCID: PMC3813907 DOI: 10.3390/toxins5101698] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/17/2013] [Accepted: 09/22/2013] [Indexed: 01/24/2023] Open
Abstract
Thirty years ago, the type 1 ribosome-inactivating protein (RIP) saporin-S6 (also known as saporin) was isolated from Saponaria officinalis L. seeds. Since then, the properties and mechanisms of action of saporin-S6 have been well characterized, and it has been widely employed in the construction of conjugates and immunotoxins for different purposes. These immunotoxins have shown many interesting results when used in cancer therapy, particularly in hematological tumors. The high enzymatic activity, stability and resistance to conjugation procedures and blood proteases make saporin-S6 a very useful tool in cancer therapy. High efficacy has been reported in clinical trials with saporin-S6-containing immunotoxins, at dosages that induced only mild and transient side effects, which were mainly fever, myalgias, hepatotoxicity, thrombocytopenia and vascular leak syndrome. Moreover, saporin-S6 triggers multiple cell death pathways, rendering impossible the selection of RIP-resistant mutants. In this review, some aspects of saporin-S6, such as the chemico-physical characteristics, the structural properties, its endocytosis, its intracellular routing and the pathogenetic mechanisms of the cell damage, are reported. In addition, the recent progress and developments of saporin-S6-containing immunotoxins in cancer immunotherapy are summarized, including in vitro and in vivo pre-clinical studies and clinical trials.
Collapse
|
17
|
Polito L, Bortolotti M, Pedrazzi M, Bolognesi A. Immunotoxins and other conjugates containing saporin-s6 for cancer therapy. Toxins (Basel) 2011; 3:697-720. [PMID: 22069735 PMCID: PMC3202841 DOI: 10.3390/toxins3060697] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 05/27/2011] [Accepted: 06/03/2011] [Indexed: 11/18/2022] Open
Abstract
Ribosome-inactivating proteins (RIPs) are a family of plant toxins that permanently damage ribosomes and possibly other cellular substrates, thus causing cell death. RIPs are mostly divided in two types: Type 1 RIPs that are single-chain enzymatic proteins, and type 2 RIPs that consist of an active A chain (similar to a type 1 RIP) linked to a B chain with lectin properties. RIP-containing conjugates have been used in many experimental strategies against cancer cells, often showing great efficacy in clinical trials. Saporin-S6, a type 1 RIP extracted from Saponaria officinalis L. seeds, has been extensively utilized to construct anti-cancer conjugates because of its high enzymatic activity, stability and resistance to conjugation procedures, resulting in the efficient killing of target cells. This review summarizes saporin-S6-containing conjugates and their application in cancer therapy, considering in-vitro and in-vivo studies both in animal models and in clinical trials. The review is structured on the basis of the targeting of hematological versus solid tumors and on the antigen recognized on the cell surface.
Collapse
Affiliation(s)
- Letizia Polito
- Department of Experimental Pathology, "Alma Mater Studiorum" University of Bologna, via San Giacomo 14, 40126-Bologna, Italy.
| | | | | | | |
Collapse
|
18
|
Li YM, Hall WA. Targeted toxins in brain tumor therapy. Toxins (Basel) 2010; 2:2645-62. [PMID: 22069569 PMCID: PMC3153175 DOI: 10.3390/toxins2112645] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 10/20/2010] [Accepted: 10/27/2010] [Indexed: 11/30/2022] Open
Abstract
Targeted toxins, also known as immunotoxins or cytotoxins, are recombinant molecules that specifically bind to cell surface receptors that are overexpressed in cancer and the toxin component kills the cell. These recombinant proteins consist of a specific antibody or ligand coupled to a protein toxin. The targeted toxins bind to a surface antigen or receptor overexpressed in tumors, such as the epidermal growth factor receptor or interleukin-13 receptor. The toxin part of the molecule in all clinically used toxins is modified from bacterial or plant toxins, fused to an antibody or carrier ligand. Targeted toxins are very effective against cancer cells resistant to radiation and chemotherapy. They are far more potent than any known chemotherapy drug. Targeted toxins have shown an acceptable profile of toxicity and safety in early clinical studies and have demonstrated evidence of a tumor response. Currently, clinical trials with some targeted toxins are complete and the final results are pending. This review summarizes the characteristics of targeted toxins and the key findings of the important clinical studies with targeted toxins in malignant brain tumor patients. Obstacles to successful treatment of malignant brain tumors include poor penetration into tumor masses, the immune response to the toxin component and cancer heterogeneity. Strategies to overcome these limitations are being pursued in the current generation of targeted toxins.
Collapse
Affiliation(s)
- Yan Michael Li
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, New York 13210, NY, USA.
| | | |
Collapse
|
19
|
Toxin-based therapeutic approaches. Toxins (Basel) 2010; 2:2519-83. [PMID: 22069564 PMCID: PMC3153180 DOI: 10.3390/toxins2112519] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 10/25/2010] [Accepted: 10/26/2010] [Indexed: 01/08/2023] Open
Abstract
Protein toxins confer a defense against predation/grazing or a superior pathogenic competence upon the producing organism. Such toxins have been perfected through evolution in poisonous animals/plants and pathogenic bacteria. Over the past five decades, a lot of effort has been invested in studying their mechanism of action, the way they contribute to pathogenicity and in the development of antidotes that neutralize their action. In parallel, many research groups turned to explore the pharmaceutical potential of such toxins when they are used to efficiently impair essential cellular processes and/or damage the integrity of their target cells. The following review summarizes major advances in the field of toxin based therapeutics and offers a comprehensive description of the mode of action of each applied toxin.
Collapse
|
20
|
Abstract
Immunotoxins are proteins used to treat cancer that are composed of an antibody fragment linked to a toxin. The immunotoxin binds to a surface antigen on a cancer cell, enters the cell by endocytosis, and kills it. The most potent immunotoxins are made from bacterial and plant toxins. Refinements over many years have produced recombinant immunotoxins; these therapeutic proteins are made using protein engineering. Individual immunotoxins are designed to treat specific cancers. To date, most success has been achieved treating hematologic tumors. Obstacles to successful treatment of solid tumors include poor penetration into tumor masses and the immune response to the toxin component of the immunotoxin, which limits the number of cycles that can be given. Strategies to overcome these limitations are being pursued.
Collapse
Affiliation(s)
- Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | |
Collapse
|
21
|
Polito L, Bolognesi A, Tazzari PL, Farini V, Lubelli C, Zinzani PL, Ricci F, Stirpe F. The conjugate Rituximab/saporin-S6 completely inhibits clonogenic growth of CD20-expressing cells and produces a synergistic toxic effect with Fludarabine. Leukemia 2004; 18:1215-22. [PMID: 15103391 DOI: 10.1038/sj.leu.2403378] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Immunotoxins are chimeric proteins consisting of a toxin coupled to an antibody. To date, several clinical trials have been conducted, and some are still ongoing, to evaluate their anti-tumor efficacy. In this view, we chemically constructed an anti-CD20 immunotoxin with the mAb Rituximab and the type 1 ribosome-inactivating protein (RIP) saporin-S6, designed for B cells non-Hodgkin's lymphoma (NHL) therapy. This immunotoxin showed a specific cytotoxicity for the CD20+ cell lines Raji and D430B, evidenced by inhibition of protein synthesis, evaluation of apoptosis and clonogenic assay. Upon conjugation, saporin-S6 increased its toxicity on target cells by at least 2 logs, with IC(50) values of 0.1-0.3 nM. The percentage of AnnexinV+ cells was over 95% in both cell lines treated with 10 nM immunotoxin. A complete elimination of Raji clones was reached with the 10 nM immunotoxin, whereas a mixture of free RIP and mAb gave about 90% of clonogenic growth. Rituximab/saporin-S6, at 10 nM concentration, also induced apoptosis in 80% of lymphoma cells from NHL patients. Moreover, sensitivity of Raji to Rituximab/saporin-S6 was augmented when cells were coincubated with Fludarabine. The synergistic toxic effect of the two drugs led to a total elimination of the neoplastic population.
Collapse
Affiliation(s)
- L Polito
- Dipartimento di Patologia Sperimentale, Università di Bologna, Bologna, Italy
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Patients with chemotherapy relapsed or refractory hematologic malignancies may be effectively treated with allogeneic or autologous stem cell transplants. However, many patients cannot be transplanted due to age, comorbidities, or lack of suitable donors. Further, a fraction of patients relapse post-transplant. Novel therapeutic agents that can kill multidrug-resistant malignant stem cells and are not myelosuppressive are needed. One class of such agents is immunotoxins. Immunotoxins consist of cell-selective ligands covalently linked to peptide toxins. The ligand delivers the molecule to specific cell surface receptors on malignant cells. The toxin triggers cell death either by reaching the cytosol and catalytically inactivating vital cell processes or by modifying the tumor cell surface membrane. We have synthesized immunotoxins for therapy of chemoresistant hematologic diseases. In this review, we will detail the synthesis of a number of these drugs and describe their preclinical and clinical activity. Several of these agents have shown dramatic antitumor effects in patients with hematologic neoplasms, and one immunotoxin has been approved for use by the US Food and Drug Administration (FDA). Over the next several decades, a growing number of these agents should reach the clinic.
Collapse
Affiliation(s)
- Arthur E Frankel
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | | | | | | |
Collapse
|
23
|
Gosselaar PH, van-Dijk AJG, de-Gast GC, Polito L, Bolognesi A, Vooijs WC, Verheul AFM, Krouwer HGJ, Marx JJM. Transferrin toxin but not transferrin receptor immunotoxin is influenced by free transferrin and iron saturation. Eur J Clin Invest 2002; 32 Suppl 1:61-9. [PMID: 11886434 DOI: 10.1046/j.1365-2362.2002.0320s1061.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Cytotoxic agents can be targeted successfully to cancer cells. The efficacy of such novel and potent anticancer strategies may be influenced by variables of iron metabolism. METHODS The in vitro cytotoxicity against glioma cells of transferrin (Tf)-based targeted toxins was compared with that of alpha-transferrin receptor (TfR)-immunotoxin. RESULTS Of four Tf-based targeted toxins, Tf-gelonin, Tf-pokeweed antiviral protein, Tf-momordin and Tf-saporin, inhibitory concentration 50% values against glioma-derived cell lines HS683 and U251, ranged from [4.8 +/- 1.5] x 10(-10) m for Tf-saporin to [26.9 +/- 15.3] x 10(-10) m for Tf-gelonin in [(3)H]-leucine incorporation assays. Tf-saporin and alpha-TfR-saporin-immunotoxin had similar efficacy, even in the more quantitative clonogenic assay (4-5 log kill with 1 x 10(-9) m) using the myeloma cell line RPMI 8226 and glioma cell line U251. However, on RPMI 8226, the efficacy of Tf-saporin 1 x 10(-9) m was reduced by 90% in the presence of 150 microg mL(-1)(=20% of normal plasma value) competing diferric transferrin, whereas the efficacy of the corresponding immunotoxin was affected only marginally. In addition, the efficacy of Tf-based conjugates will depend on their iron saturation state. Iron desaturation of Tf-saporin was demonstrated by [(59)Fe]-labelling, subsequent CM-Sepharose chromatography and SDS-PAGE. Desaturation led to virtually complete loss of affinity for the transferrin receptor, as determined by flow cytometry, which could be largely restored upon resaturation. CONCLUSION Transferrin-based toxin conjugates are strongly influenced by the presence of free transferrin and the iron saturation state. The corresponding alpha-transferrin receptor-immunotoxin does not show these disadvantages, has similar efficacy and should be preferred for further experiments.
Collapse
Affiliation(s)
- P H Gosselaar
- University Medical Centre Utrecht, Eijkman Winkler Institute G04.614, Heidelberglaan 100, PO Box 85500, 3508GA Utrecht, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lorenzetti I, Meneguzzi A, Fracasso G, Potrich C, Costantini L, Chiesa E, Legname G, Menestrina G, Tridente G, Colombatti M. Genetic grafting of membrane-acting peptides to the cytotoxin dianthin augments its ability to de-stabilize lipid bilayers and enhances its cytotoxic potential as the component of transferrin-toxin conjugates. Int J Cancer 2000; 86:582-9. [PMID: 10797275 DOI: 10.1002/(sici)1097-0215(20000515)86:4<582::aid-ijc22>3.0.co;2-i] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Three chimeric proteins were obtained by fusing together the dianthin gene and DNA fragments encoding for the following membrane-acting peptides: the N-terminus of protein G of the vesicular stomatitis virus (KFT25), the N terminus of the HA2 hemagglutinin of influenza virus (pHA2), and a membrane-acting peptide (pJVE). Chimeric dianthins (KFT25DIA, pHA2DIA and pJVEDIA) retained full enzymatic activity in cell-free assays and showed increased ability to induce pH-dependent calcein release from large unilamellar vesicles (LUVs). pHA2DIA and pJVEDIA also showed faster kinetics of interaction with LUVs, while KFT25DIA and pHA2DIA displayed a reduced cytotoxicity as compared to wild-type dianthin. Conjugates made by chemically cross-linking KFT25DIA or pJVEDIA and human transferrin (Tfn) showed greater cell-killing efficiency than conjugates of Tfn and wild-type dianthin. As a consequence, by fusion of membrane-acting peptides to the dianthin sequence the specificity factor (i.e., the ratio between non-specific and specific toxicity) of Tfn-KFT25DIA, Tfn-pHA2DIA and Tfn-pJVEDIA was increased with respect to that of Tfn-based conjugates made with wild-type dianthin. Taken together, our results suggest that genetic fusion of membrane-acting peptides to enzymatic cytotoxins results in the acquisition of new physico-chemical properties exploitable for designing new recombinant cytotoxins and to tackle cell-intoxication mechanisms.
Collapse
Affiliation(s)
- I Lorenzetti
- Section of Immunology, Department of Pathology, University of Verona, Verona, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Sforzini S, de Totero D, Gaggero A, Ippoliti R, Glennie MJ, Canevari S, Stein H, Ferrini S. Targeting of saporin to Hodgkin's lymphoma cells by anti-CD30 and anti-CD25 bispecific antibodies. Br J Haematol 1998; 102:1061-8. [PMID: 9734659 DOI: 10.1046/j.1365-2141.1998.00859.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
CD25 and CD30 represent suitable target molecules for bispecific antibody (bimAb)-driven toxin delivery to lymphoid tumour cells. We describe two new anti-CD30/anti-saporin bimAbs (termed CD30 x sap1 and CD30 x sap2), produced by hybrid hybridomas, which react against non-cross-reactive epitopes of the saporin molecule, and compared their effect with a bimAb reacting with saporin and with CD25 (CD25 x sap1). In a protein synthesis inhibition assay these bimAbs were able to enhance saporin toxicity (IC50 = 8.5 x 10(-9) M in the absence of mAbs) with a similar activity: in the presence of 10(-9) M CD30 x sap1 bimAb the IC50 was 2.75 x 10(-11) M, whereas with CD30 x sap2 bimAb the IC50 was 6.5 x 10(-11) M and CD25 x sap1 bimAb displayed an IC50 of 3 x 10(-11) M (as saporin). The combined use of the two anti-CD30 bimAbs further increased cytotoxicity by 100-fold, resulting in an IC50 of 1.9 x 10(-13) M. A slightly less efficient improvement was obtained by combining the CD25 x sap1 bimAb with the CD30 x sap2 bimAb directed against a different toxin epitope (saporin IC50 to 7 x 10(-13) M). In contrast, no synergistic effect was observed using the combination of the anti-CD25 bimAb with the anti-CD30 bimAb reacting with the same epitope of saporin (IC50 = 4.5 x 10(-11) M). Analysis of FITC-saporin binding to L540 cells by flow cytometry demonstrated that the appropriate combinations of the two anti-CD30/anti-saporin bimAbs or of the anti-CD30/anti-saporin and anti-CD25/anti-saporin bimAbs had a cooperative effect on the binding of the ribosome-inactivating protein (RIP) to the cells, when compared with single bimAbs.
Collapse
MESH Headings
- Antibodies, Bispecific/biosynthesis
- Antibodies, Bispecific/immunology
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Phytogenic/immunology
- Antineoplastic Agents, Phytogenic/pharmacokinetics
- Antineoplastic Agents, Phytogenic/pharmacology
- Cell Death/drug effects
- Dose-Response Relationship, Drug
- Hodgkin Disease/immunology
- Hodgkin Disease/metabolism
- Humans
- Hybridomas/immunology
- Immunotoxins/pharmacokinetics
- Ki-1 Antigen/immunology
- N-Glycosyl Hydrolases
- Neoplasm Proteins/biosynthesis
- Plant Proteins/immunology
- Plant Proteins/pharmacokinetics
- Plant Proteins/pharmacology
- Receptors, Interleukin-2/immunology
- Ribosome Inactivating Proteins, Type 1
- Saporins
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- S Sforzini
- Istituto Nazionale per la Ricerca sul Cancro, Centro di Biotecnologie Avanzate, Genova, Italy
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Bolognesi A, Tazzari PL, Olivieri F, Polito L, Lemoli R, Terenzi A, Pasqualucci L, Falini B, Stirpe F. Evaluation of immunotoxins containing single-chain ribosome-inactivating proteins and an anti-CD22 monoclonal antibody (OM124): in vitro and in vivo studies. Br J Haematol 1998; 101:179-88. [PMID: 9576199 DOI: 10.1046/j.1365-2141.1998.00665.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immunotoxins were prepared with three ribosome-inactivating proteins (RIP), momordin, pokeweed antiviral protein from seeds (PAP-S) and saporin-S6, linked to the anti-CD22 monoclonal antibody OM124. These immunotoxins inhibited protein synthesis by CD22-expressing cell lines Daudi, EHM, BJAB, Raji and BM21 with IC50 (concentration causing 50% inhibition) ranging from < 5 x 10(-15) to 7.6 x 10(-11) M as RIP, and IC90 (concentration causing 90% inhibition) ranging from 5 x 10(-14) to 5 x 10(-8)M, with no effect on a CD22-negative HL60 cell line at the highest concentration tested (5 x 10[-8] M). Apoptosis was induced in sensitive cells. The formation of bone marrow colonies was inhibited by no more than 40% by the immunotoxins at concentrations up to 10(-9) M. Treatment with the immunotoxins, alone or in combination, significantly extended the survival time of mice bearing transplanted Daudi cells. A treatment with cyclophosphamide and OM124/saporin immunotoxin was particularly effective in SCID mice transplanted with a low number of cells (3 x 10[-6]), when 60% of the animals remained tumour-free.
Collapse
Affiliation(s)
- A Bolognesi
- Dipartimento di Patologia Sperimentale, Università di Bologna, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ippoliti R, Lendaro E, Bellelli A, Fiani ML, Benedetti PA, Evangelista V, Brunori M. A saporin-insulin conjugate: synthesis and biochemical characterization. NATURAL TOXINS 1996; 4:156-62. [PMID: 8887945 DOI: 10.1002/19960404nt2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Saporin, a single-chain, non-cytotoxic, ribosome-inactivating protein from Saponaria officinalis, was chemically linked to the hormone insulin in a 1:1 complex. To follow by dynamic video microscopy the endocytosis and intracellular transport in vivo, a second covalent conjugate with a saporin derivative labelled with fluorescein isothiocyanate was also prepared. Both conjugates were characterized with reference to homogeneity, stoichiometry, optical spectroscopy and toxicity. Both were found to exhibit scarce toxicity toward both CHO and HEP G2 cells; optical video microscopy on living cells indicates that reduced toxicity may be (partly) due to a very limited binding of the saporin-insulin conjugate to membrane receptors. These results suggest a strategy for new possible covalent conjugates of saporin with alternative and specific macromolecular carriers.
Collapse
Affiliation(s)
- R Ippoliti
- Dipartimento di Scienze Biochimiche e Centro di Biologia Molecolare del CNR, Università La Sapienza, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
28
|
Battelli MG, Polito L, Bolognesi A, Lafleur L, Fradet Y, Stirpe F. Toxicity of ribosome-inactivating proteins-containing immunotoxins to a human bladder carcinoma cell line. Int J Cancer 1996; 65:485-90. [PMID: 8621232 DOI: 10.1002/(sici)1097-0215(19960208)65:4<485::aid-ijc16>3.0.co;2-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Immunotoxins were prepared by linking the type 1 ribosome-inactivating proteins (RIP) momordin I, pokeweed antiviral protein from seeds (PAP-S) and saporin-S6 to the 48-127 monoclonal antibody (MAb) recognising a glycoprotein (gp54) expressed on all human bladder tumours tested and on human bladder carcinoma cell lines, in particular on the T24 cell line. T24 cells required a 2 hr contact with immunotoxins to ensure binding and endocytosis. A time course of exposure, followed by further incubation without the immunotoxins, showed that maximum inhibition of protein synthesis by T24 cells was reached after 2 hr of contact followed by 3 days without the immunotoxins. Under optimal conditions, 48-127/RIP immunotoxins at nanomolar concentrations inhibited by 50% protein synthesis of target T24 cells. No toxicity was observed if (i) target cells were treated with non-conjugated RIP, (ii) target cells were treated with momordin I- or PAP-S-containing immunotoxins made with an irrelevant antibody and (iii) a non-target cell line was treated with the same 2 RIP conjugated to 48-127 antibody. The in vitro selective toxicity of these immunotoxins encourages further studies in view of a possible use in clinical trials for the local therapy of human bladder carcinomas.
Collapse
Affiliation(s)
- M G Battelli
- Department of Experimental Pathology, University of Bologna, Italy
| | | | | | | | | | | |
Collapse
|
29
|
Sforzini S, Bolognesi A, Meazza R, Marciano S, Tazzari PL, Stein H, Stirpe F, Ferrini S. Targeting of type 1 ribosome-inactivating proteins to CD30+ or CD25+ hematologic neoplasias by bispecific antibodies. JOURNAL OF HEMATOTHERAPY 1995; 4:429-32. [PMID: 8581380 DOI: 10.1089/scd.1.1995.4.429] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In this study, we compared the ability of different bispecific monoclonal antibodies (BsmAb) and immunotoxins to deliver the type 1 ribosome-inactivating proteins (RIP) saporin and gelonin through the CD25 or CD30 target molecules to Hodgkin's lymphoma cells. An anti-CD25/antisaporin and an anti-CD30/antisaporin BsmAb enhanced the toxicity of the relevant RIP against the CD25+CD30+ L540 Hodgkin's lymphoma cell line, although targeting by anti-CD30 BsmAb appeared eight times more efficient. Two anti-CD30/antigelonin BsmAb, reacting with different epitopes of the gelonin molecule, were able to enhance gelonin toxicity against L540 cells and had a synergistic effect when used in combination. Among CD25-CD30+ Hodgkin's lymphoma lines, which were resistant to targeting by anti-CD25/saporin BsmAb, one (L428) was sensitive to both gelonin and saporin delivered by anti-CD30 BsmAb. Another CD25-CD30+ cell line (COLE) was completely resistant to the toxic effect of gelonin targeted by the two synergistic BsmAb, as well as to an anti-CD30/gelonin immunotoxin. However, these cells were partially sensitive to saporin delivered by an anti-CD30/anti-saporin BsmAb, and they were efficiently killed by an anti-CD30/saporin immunotoxin. These results indicate that heterogeneity in the sensitivity to certain RIP, such as gelonin, exists among tumor cells of the same histotype.
Collapse
Affiliation(s)
- S Sforzini
- Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Sforzini S, Bolognesi A, Meazza R, Marciano S, Casalini P, Dürkop H, Tazzari PL, Stein H, Stirpe F, Ferrini S. Differential sensitivity of CD30+ neoplastic cells to gelonin delivered by anti-CD30/anti-gelonin bispecific antibodies. Br J Haematol 1995; 90:572-7. [PMID: 7646996 DOI: 10.1111/j.1365-2141.1995.tb05586.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Lymphocyte activation antigens, such as CD30, represent suitable target molecules for antibody-driven drug delivery in haemopoietic malignancies. A ribosome-inactivating protein (RIP) type 1 of potential interest for mAb targeting is gelonin, which displays a lower toxicity, as compared to other RIPs. In this study, two anti-CD30/antigelonin bispecific monoclonal antibodies (bimAbs), secreted by hybrid hybridomas, were used to deliver this RIP to CD30+ tumour cells. The two bimAbs, termed D4 and A18, were produced using the same anti-CD30 mAb and two anti-gelonin mAbs, directed to unrelated epitopes of the gelonin molecule. These bimAbs enhanced gelonin toxicity (IC50 5 x 10(-8) M, in the absence of mAbs) against the CD30+ L540 Hodgkin's lymphoma cell line in a protein synthesis inhibition assay. Thus, in the presence of 10(-9) M D4 bimAb, protein synthesis was inhibited with an IC50 of 5 x 10(-10) M as gelonin, whereas with A18 bimAb the IC50 was 8 x 10(-11) M. More interestingly, the combined use of the two bimAbs had a synergistic effect, since the IC50 of gelonin reached 6 x 10(-12) M. Among CD30 tumour cell lines, the Hodgkin's lymphoma L428 was also sensitive to gelonin delivered by bimAbs (IC50 6 x 10(-11) M), whereas the COLE Hodgkin's cell line and the T-ALL Jurkat were completely resistant to the toxic effect of gelonin and bimAbs. COLE and Jurkat cells were also resistant to a gelonin/anti-CD30 conventional immunotoxin, whereas they were sensitive to a saporin/anti-CD30 immunotoxin. This suggests that the resistance to gelonin is not related to a lack of internalization through the CD30 molecule but is associated with some property of the RIP.
Collapse
Affiliation(s)
- S Sforzini
- Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
French RR, Penney CA, Browning AC, Stirpe F, George AJ, Glennie MJ. Delivery of the ribosome-inactivating protein, gelonin, to lymphoma cells via CD22 and CD38 using bispecific antibodies. Br J Cancer 1995; 71:986-94. [PMID: 7734325 PMCID: PMC2033802 DOI: 10.1038/bjc.1995.190] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
It is well established that bispecific antibodies (BsAbs) can be used effectively in targeting the ribosome-inactivating protein (RIP), saporin, against neoplastic B cells. We have now extended this delivery system for use with gelonin. By measuring antigen-binding characteristics and epitope mapping a panel of anti-gelonin MAbs using the IAsys resonant mirror bisensor, we were able to rapidly select the most suitable for making BaAbs. The Fab' fragments from these MAbs were chemically conjugated with Fab' from either anti-CD22 or anti-CD38. Cytotoxicity assays showed that BsAbs were highly efficient at delivering gelonin to cultured Daudi cells and achieved levels of toxicity which correlated closely with the affinity of the BsAbs. Using pairs of anti-CD22 BsAbs we were able to generate bivalent BsAb-gelonin complexes which achieved IC50 values of 2 x 10(-11) M gelonin, a potency which is equivalent to that reached by saporin in this targeting system. However, because gelonin is 5-10 times less toxic than saporin, the therapeutic ratio for gelonin is superior, making it potentially a more useful agent for human treatment. Cytotoxicity assays and kinetic analysis showed that targeting gelonin via CD38 was 2-5 times less effective than delivery through CD22. However, with a pair of BsAbs designed to co-target gelonin via CD22 and CD38, the cytotoxicity achieved equalled that obtained with a pair of anti-CD22 BsAbs (IC50 = 1 x 10(-11) M). This important result suggests that the anti-CD38 helps bind the gelonin to the cell and is then 'dragged' or 'piggy-backed' into the cell by the anti-CD22 BsAb. The implication of these findings for cancer therapy is discussed.
Collapse
MESH Headings
- ADP-ribosyl Cyclase
- ADP-ribosyl Cyclase 1
- Animals
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/metabolism
- Antigens, CD/immunology
- Antigens, Differentiation/immunology
- Antigens, Differentiation, B-Lymphocyte/immunology
- Antigens, Neoplasm/immunology
- Burkitt Lymphoma/drug therapy
- Burkitt Lymphoma/immunology
- Burkitt Lymphoma/metabolism
- Cell Adhesion Molecules
- Immunotoxins/metabolism
- Immunotoxins/pharmacokinetics
- Immunotoxins/pharmacology
- Iodine Radioisotopes
- Kinetics
- Lectins
- Leucine/pharmacokinetics
- Membrane Glycoproteins
- Mice
- Mice, Inbred BALB C
- Mice, Inbred CBA
- N-Glycosyl Hydrolases
- Plant Proteins/administration & dosage
- Plant Proteins/pharmacokinetics
- Plant Proteins/toxicity
- Protein Synthesis Inhibitors/administration & dosage
- Protein Synthesis Inhibitors/pharmacokinetics
- Protein Synthesis Inhibitors/toxicity
- Ribosome Inactivating Proteins, Type 1
- Saporins
- Sialic Acid Binding Ig-like Lectin 2
- Tritium
Collapse
Affiliation(s)
- R R French
- Lymphoma Research Unit, Tanovus Laboratory, General Hospital, Southampton, UK
| | | | | | | | | | | |
Collapse
|
32
|
Bolognesi A, Tazzari PL, Legname G, Olivieri F, Modena D, Conte R, Stirpe F. Anti-CD30 immunotoxins with native and recombinant dianthin 30. Cancer Immunol Immunother 1995; 40:109-14. [PMID: 7882380 PMCID: PMC11037669 DOI: 10.1007/bf01520292] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/1994] [Accepted: 10/31/1994] [Indexed: 01/27/2023]
Abstract
Immunotoxins were prepared with a Ber-H2 (anti-CD30) monoclonal antibody and native or recombinant dianthin 30, a ribosome-inactivating protein from Dianthus caryophyllus (carnation). Both immunotoxins selectively inhibited protein synthesis by CD30+ cell lines D430B (lymphoblastoid, infected with Epstein-Barr virus), L428 and L540 (both from Hodgkin's lymphoma). IC50 values (concentrations, as dianthin, causing 50% inhibition) ranged from 324 pM to 479 pM (immunotoxin with native dianthin 30) or from 45 pM to 182 pM (immunotoxin with recombinant dianthin 30). The effect of either immunotoxin on protein synthesis by the CD30+ cell line K562 (from a chronic myeloid leukaemia) was not different from that of free dianthin (IC50 higher than nM).
Collapse
Affiliation(s)
- A Bolognesi
- Dipartimento di Patologia sperimentale dell'Università di Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
33
|
Siegall CB, Wolff EA, Gawlak SL, Paul L, Chace D, Mixan B. Immunotoxins as cancer chemotherapeutic agents. Drug Dev Res 1995. [DOI: 10.1002/ddr.430340210] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
34
|
Better M, Bernhard S, Fishwild D, Nolan P, Bauer R, Kung A, Carroll S. Gelonin analogs with engineered cysteine residues form antibody immunoconjugates with unique properties. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(17)36930-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
35
|
Barbieri L, Battelli MG, Stirpe F. Ribosome-inactivating proteins from plants. BIOCHIMICA ET BIOPHYSICA ACTA 1993; 1154:237-82. [PMID: 8280743 DOI: 10.1016/0304-4157(93)90002-6] [Citation(s) in RCA: 546] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- L Barbieri
- Dipartimento di Patologia Sperimentale, Università di Bologna, Italy
| | | | | |
Collapse
|
36
|
Kohn FR, Fishwild DM, Bernhard SL, Better M, Kung AH. Efficacy of anti-CD5 F(ab')2 and Fab' immunoconjugates in human peripheral blood lymphocyte-reconstituted severe combined immunodeficient mice. INTERNATIONAL JOURNAL OF IMMUNOPHARMACOLOGY 1993; 15:871-8. [PMID: 7504661 DOI: 10.1016/0192-0561(93)90004-i] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A human peripheral blood lymphocyte-reconstituted severe combined immunodeficient (hu-PBL-SCID) mouse model was used to compare in vivo efficacy of immunoconjugates directed against the CD5 antigen present on human T-cells. Four anti-CD5 immunoconjugates were tested, composed of chimeric human-mouse (cH65) F(ab')2 or Fab' fragments chemically linked to recombinant gelonin (rGEL) or the 30,000 M(r) glycoform of ricin A chain (RTA30). Immunoconjugate treatment was initiated approximately 3 weeks after PBL transplantation and consisted of five consecutive daily bolus i.v. injections. Efficacy was subsequently assessed by quantitation of human T-cells in spleens, blood and peritoneal lavage fluid using 3-color flow cytometry. cH65 F(ab')2- and cH65 Fab'-rGEL conjugates were essentially equally effective at depleting human T-cells from SCID mouse tissues, suggesting that bivalent binding is not required for efficacy when rGEL is the cytotoxic moiety. Treatment with unconjugated F(ab')2, unconjugated Fab' or a Fab-rGEL immunoconjugate of irrelevant binding specificity did not result in a significant depletion of T-cells, demonstrating that the cytotoxic moiety and a relevant human T-cell binding moiety are both required for efficacy. In contrast to the results observed with the rGEL conjugates, cH65 Fab'-RTA30 was not as effective as cH65 F(ab')2-RTA30 in depleting human T-cells from SCID mouse tissues. This paralleled in vitro findings in a human PBMC cytotoxicity assay, which demonstrated that cH65 Fab'-RTA30 was 17-fold less potent than cH65 F(ab')2-RTA30 and approximately 50-fold less potent than the rGEL conjugates.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- F R Kohn
- Department of Pharmacology and Toxicology, XOMA Corporation, Berkeley, CA 94710
| | | | | | | | | |
Collapse
|