1
|
Schoenfeld K, Harwardt J, Kolmar H. Better safe than sorry: dual targeting antibodies for cancer immunotherapy. Biol Chem 2024; 405:443-459. [PMID: 38297991 DOI: 10.1515/hsz-2023-0329] [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: 10/17/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
Antibody-based therapies are revolutionizing cancer treatment and experience a steady increase from preclinical and clinical pipelines to market share. While the clinical success of monoclonal antibodies is frequently limited by low response rates, treatment resistance and various other factors, multispecific antibodies open up new prospects by addressing tumor complexity as well as immune response actuation potently improving safety and efficacy. Novel antibody approaches involve simultaneous binding of two antigens on one cell implying increased specificity and reduced tumor escape for dual tumor-associated antigen targeting and enhanced and durable cytotoxic effects for dual immune cell-related antigen targeting. This article reviews antibody and cell-based therapeutics for oncology with intrinsic dual targeting of either tumor cells or immune cells. As revealed in various preclinical studies and clinical trials, dual targeting molecules are promising candidates constituting the next generation of antibody drugs for fighting cancer.
Collapse
Affiliation(s)
- Katrin Schoenfeld
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Strasse 4, D-64287 Darmstadt, Germany
| | - Julia Harwardt
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Strasse 4, D-64287 Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Strasse 4, D-64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|
2
|
Efremenko E, Aslanli A, Lyagin I. Advanced Situation with Recombinant Toxins: Diversity, Production and Application Purposes. Int J Mol Sci 2023; 24:ijms24054630. [PMID: 36902061 PMCID: PMC10003545 DOI: 10.3390/ijms24054630] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 03/04/2023] Open
Abstract
Today, the production and use of various samples of recombinant protein/polypeptide toxins is known and is actively developing. This review presents state-of-the-art in research and development of such toxins and their mechanisms of action and useful properties that have allowed them to be implemented into practice to treat various medical conditions (including oncology and chronic inflammation applications) and diseases, as well as to identify novel compounds and to detoxify them by diverse approaches (including enzyme antidotes). Special attention is given to the problems and possibilities of the toxicity control of the obtained recombinant proteins. The recombinant prions are discussed in the frame of their possible detoxification by enzymes. The review discusses the feasibility of obtaining recombinant variants of toxins in the form of protein molecules modified with fluorescent proteins, affine sequences and genetic mutations, allowing us to investigate the mechanisms of toxins' bindings to their natural receptors.
Collapse
Affiliation(s)
- Elena Efremenko
- Correspondence: ; Tel.: +7-(495)-939-3170; Fax: +7-(495)-939-5417
| | | | | |
Collapse
|
3
|
Tumor-specific intracellular delivery: peptide-guided transport of a catalytic toxin. Commun Biol 2023; 6:60. [PMID: 36650239 PMCID: PMC9845330 DOI: 10.1038/s42003-022-04385-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 12/20/2022] [Indexed: 01/19/2023] Open
Abstract
There continues to be a need for cancer-specific ligands that can deliver a wide variety of therapeutic cargos. Ligands demonstrating both tumor-specificity and the ability to mediate efficient cellular uptake of a therapeutic are critical to expand targeted therapies. We previously reported the selection of a peptide from a peptide library using a non-small cell lung cancer (NSCLC) cell line as the target. Here we optimize our lead peptide by a series of chemical modifications including truncations, N-terminal capping, and changes in valency. The resultant 10 amino acid peptide has an affinity of <40 nM on four different NSCLC cell lines as a monomer and is stable in human serum for >48 h. The peptide rapidly internalizes upon cell binding and traffics to the lysosome. The peptide homes to a tumor in an animal model and is retained up to 72 h. Importantly, we demonstrate that the peptide can deliver the cytotoxic protein saporin specifically to cancer cells in vitro and in vivo, resulting in an effective anticancer agent.
Collapse
|
4
|
Rabia E, Garambois V, Dhommée C, Larbouret C, Lajoie L, Buscail Y, Jimenez-Dominguez G, Choblet-Thery S, Liaudet-Coopman E, Cerutti M, Jarlier M, Ravel P, Gros L, Pirot N, Thibault G, Zhukovsky EA, Gérard PE, Pèlegrin A, Colinge J, Chardès T. Design and selection of optimal ErbB-targeting bispecific antibodies in pancreatic cancer. Front Immunol 2023; 14:1168444. [PMID: 37153618 PMCID: PMC10157173 DOI: 10.3389/fimmu.2023.1168444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/06/2023] [Indexed: 05/10/2023] Open
Abstract
The ErbB family of receptor tyrosine kinases is a primary target for small molecules and antibodies for pancreatic cancer treatment. Nonetheless, the current treatments for this tumor are not optimal due to lack of efficacy, resistance, or toxicity. Here, using the novel BiXAb™ tetravalent format platform, we generated bispecific antibodies against EGFR, HER2, or HER3 by considering rational epitope combinations. We then screened these bispecific antibodies and compared them with the parental single antibodies and antibody pair combinations. The screen readouts included measuring binding to the cognate receptors (mono and bispecificity), intracellular phosphorylation signaling, cell proliferation, apoptosis and receptor expression, and also immune system engagement assays (antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity). Among the 30 BiXAbs™ tested, we selected 3Patri-1Cetu-Fc, 3Patri-1Matu-Fc and 3Patri-2Trastu-Fc as lead candidates. The in vivo testing of these three highly efficient bispecific antibodies against EGFR and HER2 or HER3 in pre-clinical mouse models of pancreatic cancer showed deep antibody penetration in these dense tumors and robust tumor growth reduction. Application of such semi-rational/semi-empirical approach, which includes various immunological assays to compare pre-selected antibodies and their combinations with bispecific antibodies, represents the first attempt to identify potent bispecific antibodies against ErbB family members in pancreatic cancer.
Collapse
Affiliation(s)
- Emilia Rabia
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Véronique Garambois
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Christine Dhommée
- GICC, Groupe Innovation et Ciblage Cellulaire, Université de Tours, Tours, France
| | - Christel Larbouret
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Laurie Lajoie
- GICC, Groupe Innovation et Ciblage Cellulaire, Université de Tours, Tours, France
| | - Yoan Buscail
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
- Réseau d’Histologie Expérimentale de Montpellier, BioCampus, Université de Montpellier, UAR3426 CNRS-US09 INSERM, Montpellier, France
| | - Gabriel Jimenez-Dominguez
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Sylvie Choblet-Thery
- Plateforme Bacfly, Baculovirus et Thérapie, BioCampus, UAR3426 CNRS-US09 INSERM, Saint-Christol-Lèz Alès, France
| | - Emmanuelle Liaudet-Coopman
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Martine Cerutti
- Plateforme Bacfly, Baculovirus et Thérapie, BioCampus, UAR3426 CNRS-US09 INSERM, Saint-Christol-Lèz Alès, France
| | - Marta Jarlier
- ICM, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Patrice Ravel
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Laurent Gros
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
- CNRS, Centre National de la Recherche Scientifique, Paris, France
| | - Nelly Pirot
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
- Réseau d’Histologie Expérimentale de Montpellier, BioCampus, Université de Montpellier, UAR3426 CNRS-US09 INSERM, Montpellier, France
| | - Gilles Thibault
- GICC, Groupe Innovation et Ciblage Cellulaire, Université de Tours, Tours, France
| | - Eugene A. Zhukovsky
- Biomunex Pharmaceuticals, Incubateur Paris Biotech santé, Hopital Cochin, Paris, France
| | | | - André Pèlegrin
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Jacques Colinge
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Thierry Chardès
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
- Plateforme Bacfly, Baculovirus et Thérapie, BioCampus, UAR3426 CNRS-US09 INSERM, Saint-Christol-Lèz Alès, France
- CNRS, Centre National de la Recherche Scientifique, Paris, France
- *Correspondence: Thierry Chardès,
| |
Collapse
|
5
|
Li M, Mei S, Yang Y, Shen Y, Chen L. Strategies to mitigate the on- and off-target toxicities of recombinant immunotoxins: an antibody engineering perspective. Antib Ther 2022; 5:164-176. [PMID: 35928456 PMCID: PMC9344849 DOI: 10.1093/abt/tbac014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/14/2021] [Accepted: 06/14/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Targeted cancer therapies using immunotoxins has achieved remarkable efficacies in hematological malignancies. However, the clinical development of immunotoxins is also faced with many challenges like anti-drug antibodies and dose-limiting toxicity issues. Such a poor efficacy/safety ratio is also the major hurdle in the research and development of antibody-drug conjugates. From an antibody engineering perspective, various strategies were summarized/proposed to tackle the notorious on target off tumor toxicity issues, including passive strategy (XTENylation of immunotoxins) and active strategies (modulating the affinity and valency of the targeting moiety of immunotoxins, conditionally activating immunotoxins in the tumor microenvironments and reconstituting split toxin to reduce systemic toxicity etc.). By modulating the functional characteristics of the targeting moiety and the toxic moiety of immunotoxins, selective tumor targeting can be augmented while sparing the healthy cells in normal tissues expressing the same target of interest. If successful, the improved therapeutic index will likely help to address the dose-limiting toxicities commonly observed in the clinical trials of various immunotoxins.
Collapse
Affiliation(s)
- Mengyu Li
- Department of Postgraduate , Jiangxi University of Traditional Chinese Medicine, Nanchang, P.R. China
- Joint Graduate School , Yangtze Delta Drug Advanced Research Institute, Nantong, P.R. China
- Joint Graduate School , Yangtze Delta Pharmaceutical College, Nantong, P.R. China
| | - Sen Mei
- Biotherapeutics , Biocytogen Jiangsu Co. Ltd, Nantong, P.R. China
| | - Yi Yang
- Joint Graduate School , Yangtze Delta Drug Advanced Research Institute, Nantong, P.R. China
- Joint Graduate School , Yangtze Delta Pharmaceutical College, Nantong, P.R. China
- Institute of Innovative Medicine , Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing, P.R. China
| | - Yuelei Shen
- Joint Graduate School , Yangtze Delta Drug Advanced Research Institute, Nantong, P.R. China
- Joint Graduate School , Yangtze Delta Pharmaceutical College, Nantong, P.R. China
- Biotherapeutics , Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing, P.R. China
- Institute of Innovative Medicine , Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing, P.R. China
| | - Lei Chen
- Biotherapeutics , Biocytogen Jiangsu Co. Ltd, Nantong, P.R. China
- Biotherapeutics , Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing, P.R. China
| |
Collapse
|
6
|
Teo MYM, Ng JJC, Fong JY, Hwang JS, Song AAL, Lim RLH, In LLA. Development of a single-chain fragment variable fused-mutant HALT-1 recombinant immunotoxin against G12V mutated KRAS colorectal cancer cells. PeerJ 2021; 9:e11063. [PMID: 33959410 PMCID: PMC8053384 DOI: 10.7717/peerj.11063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 02/14/2021] [Indexed: 12/27/2022] Open
Abstract
Background KRAS oncogenes harboring codon G12 and G13 substitutions are considered gatekeeper mutations which drive oncogenesis in many cancers. To date, there are still no target-specific vaccines or drugs available against this genotype, thus reinforcing the need towards the development of targeted therapies such as immunotoxins. Methods This study aims to develop a recombinant anti-mKRAS scFv-fused mutant Hydra actinoporin-like-toxin-1 (mHALT-1) immunotoxin that is capable of recognizing and eradicating codon-12 mutated k-ras antigen abnormal cells. One G13D peptide mimotope (164-D) and one G12V peptide mimotope (68-V) were designed to elicit antigen specific IgG titres against mutated K-ras antigens in immunised Balb/c mice. The RNA was extracted from splenocytes following ELISA confirmation on post-immunized mice sera and was reverse transcribed into cDNA. The scFv combinatorial library was constructed from cDNA repertoire of variable regions of heavy chain (VH) and light chain (VL) fusions connected by a flexible glycine-serine linker, using splicing by overlap extension PCR (SOE-PCR). Anti-mKRAS G12V and G13D scFvs were cloned in pCANTAB5E phagemid and superinfected with helper phage. After few rounds of bio-panning, a specific mKRAS G12V and G13D scFv antibody against G12V and G13D control mimotope was identified and confirmed using ELISA without any cross-reactivity with other mimotopes or controls. Subsequently, the anti-mKRAS scFv was fused to mHALT-1 using SOE-PCR and cloned in pET22b vector. Expressed recombinant immunotoxins were analyzed for their effects on cell proliferation by the MTT assay and targeted specificity by cell-based ELISA on KRAS-positive and KRAS-negative cancer cells. Results The VH and VL genes from spleen RNA of mice immunized with 164-D and 68-V were amplified and randomly linked together, using SOE-PCR producing band sizes about 750 bp. Anti-mKRAS G12V and G13D scFvs were constructed in phagemid pCANTAB5E vectors with a library containing 3.4 × 106 and 2.9 × 106 individual clones, respectively. After three rounds of bio-panning, the anti-mKRAS G12V-34 scFv antibody against G12V control mimotope was identified and confirmed without any cross-reactivity with other controls using ELISA. Anti-mKRAS G12V-34 scFv fragment was fused to mHALT-1 toxin and cloned in pET22b vector with expression as inclusion bodies in E. coli BL21(DE3) (molecular weight of ~46.8 kDa). After successful solubilization and refolding, the mHALT-1-scFv immunotoxin exhibited cytotoxic effects on SW-480 colorectal cancer cells with IC50 of 25.39 μg/mL, with minimal cytotoxicity effect on NHDF cells. Discussion These results suggested that the development of such immunotoxins is potentially useful as an immunotherapeutic application against KRAS-positive malignancies.
Collapse
Affiliation(s)
- Michelle Yee Mun Teo
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Jeremy Jeack Ceen Ng
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Jung Yin Fong
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Jung Shan Hwang
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Adelene Ai-Lian Song
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Renee Lay Hong Lim
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Lionel Lian Aun In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Wilayah Persekutuan Kuala Lumpur, Malaysia
| |
Collapse
|
7
|
Taghizadegan N, Firoozrai M, Nassiri M, Ariannejad H. A novel strategy for engineering of a smart generation of immune ribonucleases against EGFR + cells. J Cell Physiol 2021; 236:4303-4312. [PMID: 33421131 DOI: 10.1002/jcp.30118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 10/04/2020] [Accepted: 10/06/2020] [Indexed: 12/26/2022]
Abstract
The overexpression of epidermal growth factor receptor (EGFR) could result in the development of solid tumors of prostate, breast, gastric, colorectal, ovarian, and head and neck, leading to carcinoma. Antibody therapies are ideal methods to overcome malignant diseases. However, immunoribonucleases are a new generation of antibodies in which an RNase binds to a specific antibody and shows a stronger ability to terminate cancer cells. In this study, we engineered Rana pipiens RNase to bind to the scFv of human antiepidermal growth factor receptor antibody. The molecular dynamic simulations confirmed protein stability and the ability of scFv-ranpirnase (rantoxin) to bind to epidermal growth factor receptor protein. Then, the rantoxin construct was synthesized in a pCDNA 3.1 Neo vector. CHO-K1 cells were used as expression hosts and the construct was transfected. Cells were selected by antibiotic therapies using neomycin, 120 mg/ml, and the high-yield colony was screened by real-time polymerase chain reaction (PCR) methods. Then, the recombinant protein production was confirmed using the sodium dodecyl sulfate polyacrylamide gel electrophoresis and western blot analyses. The molecular dynamic simulation (MDS) confirmed that the I467, S468, Q408, and H409 amino acids of EGFR bonded well to rantoxin. As revealed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and western blot analyses, the rantoxin production and PCR analysis showed that the T3 colony can produce rantoxin messenger RNA fourfold higher than the GAPDH gene. The immunotoxin function was assessed in A431 cancer cells and EGFR-negative HEK293 cells, and IC50 values were estimated to be 22.4 ± 3 and >620.4 ± 5 nM, respectively. The results indicated that the immunotoxins produced in this study had the potential for use as anticancer drugs.
Collapse
Affiliation(s)
- Nooshin Taghizadegan
- Department of Biochemistry, Shahrood Branch, Islamic Azad University, Shahrood, Iran
| | - Mohsen Firoozrai
- Department of Biochemistry, Shahrood Branch, Islamic Azad University, Shahrood, Iran
| | | | - Hamid Ariannejad
- Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
8
|
Guo R, Yang Y, Zhang D, Du J, Zhu X, Liu Y, Yang F, Lin J. A bispecific immunotoxin (IHPP) with a long half-life targeting HER2 and PDGFRβ exhibited improved efficacy against HER2-positive tumors in a mouse xenograft model. Int J Pharm 2021; 592:120037. [PMID: 33161038 DOI: 10.1016/j.ijpharm.2020.120037] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/07/2020] [Accepted: 10/28/2020] [Indexed: 12/27/2022]
Abstract
Multiple signaling pathways are usually involved in the development of tumors. Compared with monospecific antibodies, bispecific antibodies can recognize two different antigens at the same time, so they are more suitable for treating tumor diseases with complex etiology. Immunotoxins have good antitumor activity, however, single targeting limits their effectiveness. Herein, we designed a Pseudomonas exotoxin A (PE)-based bispecific immunotoxin IgBD-HER2-PDGFRβ-PE38 which could distinguish HER2 and PDGFRβ target in tumor. Meanwhile, IgG-affinity could extend the serum retention of immunotoxins after in vivo injection. In this work, we first detected the selective binding of the immunotoxins and antitumor effect in vitro. Compared with control group, IgBD-HER2-PDGFRβ-PE38 exhibited improved efficacy against HER2-positive tumors in an NCI-N87 subcutaneous xenograft model. Then, transcriptome sequencing was performed on tumor tissue originating from different treatment groups of mice bearing NCI-N87 tumors. Seven significantly differentially expressed genes were screened based on human genes, and the differential mouse genes were enriched based on the Reactome Pathway Database. At last, the RNA sequencing results were verified by real-time PCR and ELISA. Therefore, the new construct bispecific immunotoxin represents a potentially attractive therapeutic modality, and the proposed strategy make them promising for use in the development of anti-HER2 cancer therapeutics.
Collapse
Affiliation(s)
- Rui Guo
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453003, Henan Province, China; Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Yun Yang
- College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Di Zhang
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453003, Henan Province, China; Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Jiang Du
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453003, Henan Province, China; Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Xinxing Zhu
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453003, Henan Province, China; Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Yanli Liu
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, Henan Province, China; College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Fen Yang
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453003, Henan Province, China; Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Juntang Lin
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453003, Henan Province, China; Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, Henan Province, China.
| |
Collapse
|
9
|
Huang S, van Duijnhoven SMJ, Sijts AJAM, van Elsas A. Bispecific antibodies targeting dual tumor-associated antigens in cancer therapy. J Cancer Res Clin Oncol 2020; 146:3111-3122. [PMID: 32989604 PMCID: PMC7679314 DOI: 10.1007/s00432-020-03404-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Bispecific antibodies (BsAbs) have emerged as a leading drug class for cancer therapy and are becoming increasingly of interest for therapeutic applications. As of April 2020, over 123 BsAbs are under clinical evaluation for use in oncology (including the two marketed BsAbs Blinatumomab and Catumaxomab). The majority (82 of 123) of BsAbs under clinical evaluation can be categorized as bispecific immune cell engager whereas a second less well-discussed subclass of BsAbs targets two tumor-associated antigens (TAAs). In this review, we summarize the clinical development of dual TAAs targeting BsAbs and provide an overview of critical considerations when designing dual TAA targeting BsAbs. METHODS Herein the relevant literature and clinical trials published in English until April 1st 2020 were searched using PubMed and ClinicalTrials.gov database. BsAbs were considered to be active in clinic if their clinical trials were not terminated, withdrawn or completed before 2018 without reporting results. Data missed by searching ClinicalTrials.gov was manually curated. RESULTS Dual TAAs targeting BsAbs offer several advantages including increased tumor selectivity, potential to concurrently modulate two functional pathways in the tumor cell and may yield improved payload delivery. CONCLUSIONS Dual TAAs targeting BsAbs represent a valuable class of biologics and early stage clinical studies have demonstrated promising anti-tumor efficacy in both hematologic malignancies and solid tumors.
Collapse
Affiliation(s)
- Shuyu Huang
- Aduro Biotech Europe, Oss, The Netherlands
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | | | - Alice J A M Sijts
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | | |
Collapse
|
10
|
Hamamichi S, Fukuhara T, Hattori N. Immunotoxin Screening System: A Rapid and Direct Approach to Obtain Functional Antibodies with Internalization Capacities. Toxins (Basel) 2020; 12:toxins12100658. [PMID: 33076544 PMCID: PMC7602748 DOI: 10.3390/toxins12100658] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/24/2022] Open
Abstract
Toxins, while harmful and potentially lethal, have been engineered to develop potent therapeutics including cytotoxins and immunotoxins (ITs), which are modalities with highly selective targeting capabilities. Currently, three cytotoxins and IT are FDA-approved for treatment of multiple forms of hematological cancer, and additional ITs are tested in the clinical trials or at the preclinical level. For next generation of ITs, as well as antibody-mediated drug delivery systems, specific targeting by monoclonal antibodies is critical to enhance efficacies and reduce side effects, and this methodological field remains open to discover potent therapeutic monoclonal antibodies. Here, we describe our application of engineered toxin termed a cell-based IT screening system. This unique screening strategy offers the following advantages: (1) identification of monoclonal antibodies that recognize cell-surface molecules, (2) selection of the antibodies that are internalized into the cells, (3) selection of the antibodies that induce cytotoxicity since they are linked with toxins, and (4) determination of state-specific activities of the antibodies by differential screening under multiple experimental conditions. Since the functional monoclonal antibodies with internalization capacities have been identified successfully, we have pursued their subsequent modifications beyond antibody drug conjugates, resulting in development of immunoliposomes. Collectively, this screening system by using engineered toxin is a versatile platform, which enables straight-forward and rapid selection for discovery of novel functional antibodies.
Collapse
Affiliation(s)
- Shusei Hamamichi
- Research Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo 113-8421, Japan;
| | - Takeshi Fukuhara
- Department of Neurology, Juntendo University School of Medicine, Tokyo 113-8421, Japan;
- Department of Research for Parkinson’s Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, Saitama 351-0198, Japan
- Correspondence: ; Tel.: +81-3-5802-2731; Fax: +81-3-5800-0547
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo 113-8421, Japan;
- Department of Research for Parkinson’s Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| |
Collapse
|
11
|
Development of Glypican-3 Targeting Immunotoxins for the Treatment of Liver Cancer: An Update. Biomolecules 2020; 10:biom10060934. [PMID: 32575752 PMCID: PMC7356171 DOI: 10.3390/biom10060934] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) accounts for most liver cancers and represents one of the deadliest cancers in the world. Despite the global demand for liver cancer treatments, there remain few options available. The U.S. Food and Drug Administration (FDA) recently approved Lumoxiti, a CD22-targeting immunotoxin, as a treatment for patients with hairy cell leukemia. This approval helps to demonstrate the potential role that immunotoxins can play in the cancer therapeutics pipeline. However, concerns have been raised about the use of immunotoxins, including their high immunogenicity and short half-life, in particular for treating solid tumors such as liver cancer. This review provides an overview of recent efforts to develop a glypican-3 (GPC3) targeting immunotoxin for treating HCC, including strategies to deimmunize immunotoxins by removing B- or T-cell epitopes on the bacterial toxin and to improve the serum half-life of immunotoxins by incorporating an albumin binding domain.
Collapse
|
12
|
Borgatti A, Dickerson EB, Lawrence J. Emerging therapeutic approaches for canine sarcomas: Pushing the boundaries beyond the conventional. Vet Comp Oncol 2019; 18:9-24. [PMID: 31749286 DOI: 10.1111/vco.12554] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/21/2022]
Abstract
Sarcomas represent a group of genomically chaotic, highly heterogenous tumours of mesenchymal origin with variable mutational load. Conventional therapy with surgery and radiation therapy is effective for managing small, low-grade sarcomas and remains the standard therapeutic approach. For advanced, high-grade, recurrent, or metastatic sarcomas, systemic chemotherapy provides minimal benefit, therefore, there is a drive to develop novel approaches. The discovery of "Coley's toxins" in the 19th century, and their use to stimulate the immune system supported the application of unconventional therapies for the treatment of sarcomas. While promising, this initial work was abandoned and treatment paradigm and disease course of sarcomas was largely unchanged for several decades. Exciting new therapies are currently changing treatment algorithms for advanced carcinomas and melanomas, and similar approaches are being applied to advance the field of sarcoma research. Recent discoveries in subtype-specific cancer biology and the identification of distinct molecular targets have led to the development of promising targeted strategies with remarkable potential to change the landscape of sarcoma therapy in dogs. The purpose of this review article is to describe the current standard of care and limitations as well as emerging approaches for sarcoma therapy that span many of the most active paradigms in oncologic research, including immunotherapies, checkpoint inhibitors, and drugs capable of cellular metabolic reprogramming.
Collapse
Affiliation(s)
- Antonella Borgatti
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota
| | - Erin B Dickerson
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Jessica Lawrence
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
13
|
Critical Issues in the Development of Immunotoxins for Anticancer Therapy. J Pharm Sci 2019; 109:104-115. [PMID: 31669121 DOI: 10.1016/j.xphs.2019.10.037] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/23/2019] [Accepted: 10/21/2019] [Indexed: 12/16/2022]
Abstract
Immunotoxins (ITs) are attractive anticancer modalities aimed at cancer-specific delivery of highly potent cytotoxic protein toxins. An IT consists of a targeting domain (an antibody, cytokine, or another cell-binding protein) chemically conjugated or recombinantly fused to a highly cytotoxic payload (a bacterial and plant toxin or human cytotoxic protein). The mode of action of ITs is killing designated cancer cells through the effector function of toxins in the cytosol after cellular internalization via the targeted cell-specific receptor-mediated endocytosis. Although numerous ITs of diverse structures have been tested in the past decades, only 3 ITs-denileukin diftitox, tagraxofusp, and moxetumomab pasudotox-have been clinically approved for treating hematological cancers. No ITs against solid tumors have been approved for clinical use. In this review, we discuss critical research and development issues associated with ITs that limit their clinical success as well as strategies to overcome these obstacles. The issues include off-target and on-target toxicities, immunogenicity, human cytotoxic proteins, antigen target selection, cytosolic delivery efficacy, solid-tumor targeting, and developability. To realize the therapeutic promise of ITs, novel strategies for safe and effective cytosolic delivery into designated tumors, including solid tumors, are urgently needed.
Collapse
|
14
|
Shafiee F, Aucoin MG, Jahanian-Najafabadi A. Targeted Diphtheria Toxin-Based Therapy: A Review Article. Front Microbiol 2019; 10:2340. [PMID: 31681205 PMCID: PMC6813239 DOI: 10.3389/fmicb.2019.02340] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 09/25/2019] [Indexed: 12/26/2022] Open
Abstract
Cancer remains one of the leading causes of death worldwide. Conventional therapeutic strategies usually offer limited specificity, resulting in severe side effects and toxicity to normal tissues. Targeted cancer therapy, on the other hand, can improve the therapeutic potential of anti-cancer agents and decrease unwanted side effects. Targeted applications of cytolethal bacterial toxins have been found to be especially useful for the specific eradication of cancer cells. Targeting is either mediated by peptides or by protein-targeting moieties, such as antibodies, antibody fragments, cell-penetrating peptides (CPPs), growth factors, or cytokines. Together with a toxin domain, these molecules are more commonly referred to as immunotoxins. Targeting can also be achieved through gene delivery and cell-specific expression of a toxin. Of the available cytolethal toxins, diphtheria toxin (DT) is one of the most frequently used for these strategies. Of the many DT-based therapeutic strategies investigated to date, two immunotoxins, OntakTM and TagraxofuspTM, have gained FDA approval for clinical application. Despite some success with immunotoxins, suicide-gene therapy strategies, whereby controlled tumor-specific expression of DT is used for the eradication of malignant cells, are gaining prominence. The first part of this review focuses on DT-based immunotoxins, and it then discusses recent developments in tumor-specific expression of DT.
Collapse
Affiliation(s)
- Fatemeh Shafiee
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marc G Aucoin
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Ali Jahanian-Najafabadi
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
15
|
Ng TJ, Teo MYM, Liew DS, Effiong PE, Hwang JS, Lim CSY, In LLA. Cytotoxic and apoptosis-inducing effects of wildtype and mutated Hydra actinoporin-like toxin 1 (HALT-1) on various cancer cell lines. PeerJ 2019; 7:e6639. [PMID: 31106043 PMCID: PMC6500716 DOI: 10.7717/peerj.6639] [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: 09/26/2018] [Accepted: 02/18/2019] [Indexed: 12/25/2022] Open
Abstract
Background Hydra actinoporin like toxin -1 (HALT-1), is a small 18.5 kDa pore forming toxin derived from Hydra magnipapillata which has been shown to elicit strong haemolytic and cytolytic activity when in contact with cell membranes. Due to its cytotoxic potency, HALT-1 was further investigated for its potential as a toxin moiety candidate in immunotoxin developmental efforts, ideally as a form of targeted therapy against cancer. Methods In this study, wtHALT-1 (wild type) and its Y110A mutated binding domain counterpart (mHALT-1) were produced and evaluated for their cytotoxic and apoptotic effects on various cancer cell lines. A total of seven different tumour and non-tumour cell lines including HeLa, HepG2, SW-620, MCF-7, CCD841CoN, NHDF and HCT116 were used. Immunofluorescence assays were used to observe membrane binding and localization changes between both HALT-1 recombinant proteins based on 6xHis-tag detection. Result Based on MTT data, mHALT-1 demonstrated a significant reduction of 82% ± 12.21% in cytotoxic activity across all cell lines after the membrane recognition domain had been mutated in comparison to the wtHALT-1. Annexin V FITC/PI assay data also indicated that HeLa, HepG2 and MCF-7 demonstrated an apoptosis-mediated cell death after being treated with wtHALT-1. Additionally, a notable difference between wtHALT-1 and mHALT-1 binding affinity was clearly observed where emission of green fluorescence along the cell membrane was observed only in wtHALT-1 treated cells. Discussion These results suggest that mHALT-1 (Y110A) can be potentially developed as a toxin-moiety candidate for the development of future immunotoxins against various human cell-based diseases.
Collapse
Affiliation(s)
- Teng Jia Ng
- Department of Biotechnology, Faculty of Applied Sciences, UCSI university, Cheras, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Michelle Yee Mun Teo
- Department of Biotechnology, Faculty of Applied Sciences, UCSI university, Cheras, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Dek Shen Liew
- Department of Biotechnology, Faculty of Applied Sciences, UCSI university, Cheras, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Paul Etim Effiong
- Department of Biotechnology, Faculty of Applied Sciences, UCSI university, Cheras, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Jung Shan Hwang
- Department of Medical Sciences, School of Healthcare and Medical Sciences, Sunway University, Subang Jaya, Selangor, Malaysia
| | - Crystale Siew Ying Lim
- Department of Biotechnology, Faculty of Applied Sciences, UCSI university, Cheras, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Lionel L A In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI university, Cheras, Wilayah Persekutuan Kuala Lumpur, Malaysia
| |
Collapse
|
16
|
Busato D, Mossenta M, Baboci L, Di Cintio F, Toffoli G, Dal Bo M. Novel immunotherapeutic approaches for hepatocellular carcinoma treatment. Expert Rev Clin Pharmacol 2019; 12:453-470. [PMID: 30907177 DOI: 10.1080/17512433.2019.1598859] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The introduction of immune checkpoint inhibitors has been lately proposed for the treatment of hepatocellular carcinoma (HCC) with respect to other cancer types. Several immunotherapeutic approaches are now under evaluation for HCC treatment including: i) antibodies acting as immune checkpoint inhibitors; ii) antibodies targeting specific tumor-associated antigens; iii) chimeric antigen receptor redirected T (CAR-T) cells targeting specific tumor-associated antigens; iv) vaccination strategies with tumor-specific epitopes. Areas covered: The review provides a wide description of the clinical trials investigating the efficacy of the main immunotherapeutic approaches proposed for the treatment of patients affected by HCC. Expert opinion: The balancing between immunostimulative and immunosuppressive factors in the context of HCC tumor microenvironment results in heterogeneous response rates to immunotherapeutic approaches such as checkpoint inhibitors, among HCC patients. In this context, it becomes crucial the identification of predictive factors determining the treatment response. A multiple approach using different biomarkers could be useful to identify the subgroup of HCC patients responsive to the treatment with a checkpoint inhibitor (as an example, nivolumab) as single agent, and to identify those patients in which other treatment regimens, such as the combination with sorafenib, or with locoregional therapies, could be more effective.
Collapse
Affiliation(s)
- Davide Busato
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy.,b Department of Life Sciences , University of Trieste , Trieste , Italy
| | - Monica Mossenta
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy.,b Department of Life Sciences , University of Trieste , Trieste , Italy
| | - Lorena Baboci
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy
| | - Federica Di Cintio
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy.,b Department of Life Sciences , University of Trieste , Trieste , Italy
| | - Giuseppe Toffoli
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy
| | - Michele Dal Bo
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy
| |
Collapse
|
17
|
Vallera DA, Kreitman RJ. Immunotoxins Targeting B cell Malignancy-Progress and Problems With Immunogenicity. Biomedicines 2018; 7:biomedicines7010001. [PMID: 30577664 PMCID: PMC6466112 DOI: 10.3390/biomedicines7010001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 01/26/2023] Open
Abstract
Few immunotoxins or targeted toxins have become mainline cancer therapies. Still immunotoxins continue to be of major interest and subject of research and development as alternative therapies for drug resistant cancer. A major matter of concern continues to be immunogenicity exemplified by the anti-toxin response of the treated patient. Since some of our most effective toxins are bacterial in nature and bacterial proteins are highly immunogenic, this review describes some efforts to address this pressing issue.
Collapse
Affiliation(s)
- Daniel A Vallera
- Laboratory of Molecular Cancer Therapeutics, Masonic Cancer Center, Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Robert J Kreitman
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
18
|
Yap WY, Hwang JS. Response of Cellular Innate Immunity to Cnidarian Pore-Forming Toxins. Molecules 2018; 23:E2537. [PMID: 30287801 PMCID: PMC6222686 DOI: 10.3390/molecules23102537] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 09/24/2018] [Accepted: 09/28/2018] [Indexed: 12/11/2022] Open
Abstract
A group of stable, water-soluble and membrane-bound proteins constitute the pore forming toxins (PFTs) in cnidarians. They interact with membranes to physically alter the membrane structure and permeability, resulting in the formation of pores. These lesions on the plasma membrane causes an imbalance of cellular ionic gradients, resulting in swelling of the cell and eventually its rupture. Of all cnidarian PFTs, actinoporins are by far the best studied subgroup with established knowledge of their molecular structure and their mode of pore-forming action. However, the current view of necrotic action by actinoporins may not be the only mechanism that induces cell death since there is increasing evidence showing that pore-forming toxins can induce either necrosis or apoptosis in a cell-type, receptor and dose-dependent manner. In this review, we focus on the response of the cellular immune system to the cnidarian pore-forming toxins and the signaling pathways that might be involved in these cellular responses. Since PFTs represent potential candidates for targeted toxin therapy for the treatment of numerous cancers, we also address the challenge to overcoming the immunogenicity of these toxins when used as therapeutics.
Collapse
Affiliation(s)
- Wei Yuen Yap
- Department of Biological Sciences, School of Science and Technology, Sunway University, No. 5 Jalan Universiti, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| | - Jung Shan Hwang
- Department of Medical Sciences, School of Healthcare and Medical Sciences, Sunway University, No. 5 Jalan Universiti, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| |
Collapse
|