1
|
Golichenari B, Heiat M, Rezaei E, Ramshini A, Sahebkar A, Gholipour N. Compromising the immunogenicity of diphtheria toxin-based immunotoxins through epitope engineering: An in silico approach. J Pharmacol Toxicol Methods 2025; 131:107571. [PMID: 39693813 DOI: 10.1016/j.vascn.2024.107571] [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: 09/07/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
Immunotoxins are genetically engineered recombinant proteins consisting of a targeting moiety, such as an antibody, and a cytotoxic toxin moiety of microbial origin. Pseudomonas exotoxin A and diphtheria toxin (DT) have been abundantly used in immunotoxins, with the latter applied as the toxin moiety of the FDA-approved drug Denileukin diftitox (ONTAK®). However, the use of immunotoxins provokes an adverse immune response in the host body against the toxin moiety, limiting their efficacy. In silico approaches have received increasing attention in protein engineering. In this study, the epitopes responsible for immunogenicity were identified through multiple platforms. By subtracting conserved and ligand-binding residues, K33, T111, and E112 were identified as common epitopes across all platforms. Substitution analysis evaluated alternative residues regarding their impact on protein stability, considering 19 different amino acid substitutions. Among the mutants explored, the T111A-E112G mutant exhibited the most destabilizing substitution for DT, thereby reducing immunogenicity. Finally, a 3D model of the mutant was generated and verified. The model was then docked with its native ligand NADH, and the complex's molecular behavior was simulated using molecular dynamics.
Collapse
Affiliation(s)
- Behrouz Golichenari
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Disease (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ehsan Rezaei
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amirreza Ramshini
- Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Nazila Gholipour
- Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Obozina AS, Pakhomov AA, Frolova AY, Deyev SM, Shipunova VO. Optimizing combination targeted immunotoxin therapy: Insights from HER2 and EpCAM expression profiles. Biochem Biophys Res Commun 2025; 746:151218. [PMID: 39752974 DOI: 10.1016/j.bbrc.2024.151218] [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: 12/04/2024] [Accepted: 12/19/2024] [Indexed: 01/15/2025]
Abstract
Molecular targeted cancer therapy is a rapidly developing field, driving progress toward greater treatment efficacy. However, targeted monotherapy often fails due to the development of multidrug resistance in tumors. The combination of multiple targeted agents emerges as a possible solution to enhance treatment outcomes by activating different signaling pathways. This study systematically investigates the combined effect of targeted agents for the oncomarkers HER2 and EpCAM on cancer cells. Specifically, the study examined the impact of anti-HER2 (DARP_9.29-LoPE) and anti-EpCAM (DARP_EC1-LoPE) immunotoxins on a panel of cancer cells expressing various levels of HER2 and EpCAM. Using the Chou-Talalay combination indices, the study revealed that cells with low HER2 expression and high EpCAM expression are not optimal targets for combined HER2/EpCAM therapy. In contrast, the most effective approach involves the usage of an equimolar ratio of immunotoxins for cells exhibiting high HER2 and moderate EpCAM expression, resulting in a synergistic therapeutic effect. These findings provide significant insights into optimizing combination anti-HER2/EpCAM therapies and hold promise for the development of more effective cancer treatment strategies.
Collapse
Affiliation(s)
- A S Obozina
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia
| | - A A Pakhomov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997, Moscow, Russia
| | - A Yu Frolova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997, Moscow, Russia
| | - S M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997, Moscow, Russia
| | - V O Shipunova
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997, Moscow, Russia.
| |
Collapse
|
3
|
Hessami A, Mogharari Z, Rahim F, Khalesi B, Jamal Nassrullah O, Reza Rahbar M, Khalili S, Jahangiri A. In silico design of a novel hybrid epitope-based antigen harboring highly exposed immunogenic peptides of BamA, OmpA, and Omp34 against Acinetobacter baumannii. Int Immunopharmacol 2024; 142:113066. [PMID: 39241518 DOI: 10.1016/j.intimp.2024.113066] [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: 05/31/2024] [Revised: 08/07/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
Acinetobacter baumannii, is among the highest priority bacteria according to the WHO categorization which necessitate the exploration of alternative strategies such as vaccination. OmpA, BamA, and Omp34 are assigned as appropriate antigens to serve in vaccine development against this pathogen. Experimentally validated exposed epitopes of OmpA and Omp34 along with selected exposed epitopes predicted by an integrative in silico approach were represented by the barrel domain of BamA as a scaffold. Among the 8 external loops of BamA, 5 loops were replaced with selected loops of OmpA and Omp34. The designed antigen was analyzed regarding the physicochemical properties, antigenicity, epitope retrieval, topology, structure, and safety. BamA is a two-domain OMP with a 16-stranded barrel in which L4, L6, and L7 were the longest loops of BamA in order. The designed antigen consisted of 478 amino acids with antigen probability of 0.7793. The novel antigen was a 16-stranded barrel. No identical 8-meric peptides were found in the human proteome against the designed antigen sequence. The designed construct was safe regarding the allergenicity, toxicity, and human proteome reactivity. The designed antigen could develop higher protection against A. baumannii in comparison to either OmpA, BamA, or Omp34 alone.
Collapse
Affiliation(s)
- Anahita Hessami
- School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Fatemeh Rahim
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares, P.O. Box: 14115-154, Tehran, Iran
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization, Karaj, Iran
| | | | - Mohammad Reza Rahbar
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Abolfazl Jahangiri
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Oghalaie A, Hosseini ME, Hosseininejad-Chafi M, Eftekhari Z, Behdani M, Kazemi-Lomedasht F. Advances in immunotoxin engineering: precision therapeutic strategies in modern oncology. Med Oncol 2024; 41:239. [PMID: 39230639 DOI: 10.1007/s12032-024-02478-3] [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] [Accepted: 08/12/2024] [Indexed: 09/05/2024]
Abstract
Immunotoxins (ITs) are specialized therapeutic agents designed for targeted treatment, particularly in cancer therapy. They consist of a monoclonal antibody or antibody fragment linked to a potent cytotoxic agent, such as bacterial- or plant-derived toxins like diphtheria toxin, ricin, or pseudomonas exotoxin. The monoclonal antibody component specifically binds to antigens expressed on the surface of target cells, facilitating the internalization of the IT. Once inside the cell, the cytotoxic agent is released, disrupting essential cellular processes and leading to cell death. This targeted approach minimizes damage to healthy tissues while effectively eliminating diseased cells. The production of ITs involves two primary methods: recombinant fusion and chemical conjugation. In recombinant fusion, genetic engineering is used to create a fusion protein that combines the antibody and toxin, ensuring precise control over their ratio and functionality. In chemical conjugation, pre-existing antibodies are chemically linked to toxins, allowing for greater flexibility in combining different antibodies and cytotoxic agents. Each method has its advantages and challenges, influencing the specificity, production complexity, and therapeutic potential of the resulting ITs. As research advances, ITs continue to show promise not only in oncology but also in treating other diseases, including inflammatory conditions and atherosclerosis. The precise targeting and potent effects of ITs make them a valuable tool in the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Akbar Oghalaie
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mahmoud Eshagh Hosseini
- Gastroenterology and Liver Department, Amiralam Hospital, University of Medical Sciences, Tehran, Iran
| | - Mohammad Hosseininejad-Chafi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Zohre Eftekhari
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
5
|
Dicovitsky RH, Schappa JT, Schulte AJ, Lang HP, Kuerbitz E, Roberts S, DePauw TA, Lewellen M, Winter AL, Stuebner K, Buettner M, Reid K, Bergsrud K, Pracht S, Chehadeh A, Feiock C, O’Sullivan MG, Carlson T, Armstrong AR, Meritet D, Henson MS, Weigel BJ, Modiano JF, Borgatti A, Vallera DA. Toxicity Profile of eBAT, a Bispecific Ligand-Targeted Toxin Directed to EGFR and uPAR, in Mice and a Clinical Dog Model. Toxins (Basel) 2024; 16:376. [PMID: 39330834 PMCID: PMC11436214 DOI: 10.3390/toxins16090376] [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: 04/25/2024] [Revised: 07/15/2024] [Accepted: 08/20/2024] [Indexed: 09/28/2024] Open
Abstract
EGFR-targeted therapies are efficacious, but toxicity is common and can be severe. Urokinase type plasminogen activator receptor (uPAR)-targeted drugs are only emerging, so neither their efficacy nor toxicity is fully established. Recombinant eBAT was created by combining cytokines EGF and uPA on the same single-chain molecule with truncated Pseudomonas toxin. Its purpose was to simultaneously target tumors and their vasculature in the tumor microenvironment. In prior studies on mice and dogs, the drug proved efficacious. Here, we report the safety of eBAT in normal wildtype, uPAR knockout, and immunoreplete and immunodeficient tumor-bearing mice, as well as in dogs with spontaneous sarcoma that more closely mirror human cancer onset. In immunocompetent mice, tumor-bearing mice, uPAR knockout mice, and mice receiving species-optimized eBAT, toxicities were mild and self-limiting. Likewise, in dogs with life-threatening sarcoma given dosages found to be biologically active, eBAT was well tolerated. In mice receiving higher doses, eBAT was associated with dose-dependent evidence of liver injury, including portal biliary hyperplasia, oval cell proliferation, lymphoplasmacytic inflammation, periportal hepatocellular microvesicular change, hemorrhage, necrosis, and apoptosis. The results support continuing the clinical development of eBAT as a therapeutic agent for individuals with sarcoma and other cancers.
Collapse
Affiliation(s)
- Rose H. Dicovitsky
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (R.H.D.); (J.T.S.); (A.J.S.); (H.P.L.); (E.K.); (S.R.); (T.A.D.); (M.L.); (C.F.); (A.R.A.); (M.S.H.); (J.F.M.); (A.B.)
| | - Jill T. Schappa
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (R.H.D.); (J.T.S.); (A.J.S.); (H.P.L.); (E.K.); (S.R.); (T.A.D.); (M.L.); (C.F.); (A.R.A.); (M.S.H.); (J.F.M.); (A.B.)
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Experimental Surgical Services, Department of Surgery, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ashley J. Schulte
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (R.H.D.); (J.T.S.); (A.J.S.); (H.P.L.); (E.K.); (S.R.); (T.A.D.); (M.L.); (C.F.); (A.R.A.); (M.S.H.); (J.F.M.); (A.B.)
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Haeree P. Lang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (R.H.D.); (J.T.S.); (A.J.S.); (H.P.L.); (E.K.); (S.R.); (T.A.D.); (M.L.); (C.F.); (A.R.A.); (M.S.H.); (J.F.M.); (A.B.)
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Comparative Molecular Biosciences Graduate Program and DVM-PhD Dual Degree Program, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
| | - Ellen Kuerbitz
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (R.H.D.); (J.T.S.); (A.J.S.); (H.P.L.); (E.K.); (S.R.); (T.A.D.); (M.L.); (C.F.); (A.R.A.); (M.S.H.); (J.F.M.); (A.B.)
| | - Sarah Roberts
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (R.H.D.); (J.T.S.); (A.J.S.); (H.P.L.); (E.K.); (S.R.); (T.A.D.); (M.L.); (C.F.); (A.R.A.); (M.S.H.); (J.F.M.); (A.B.)
| | - Taylor A. DePauw
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (R.H.D.); (J.T.S.); (A.J.S.); (H.P.L.); (E.K.); (S.R.); (T.A.D.); (M.L.); (C.F.); (A.R.A.); (M.S.H.); (J.F.M.); (A.B.)
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Microbiology, Immunology, and Cancer Biology Graduate Program, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mitzi Lewellen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (R.H.D.); (J.T.S.); (A.J.S.); (H.P.L.); (E.K.); (S.R.); (T.A.D.); (M.L.); (C.F.); (A.R.A.); (M.S.H.); (J.F.M.); (A.B.)
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Amber L. Winter
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Clinical Investigation Center, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
| | - Kathy Stuebner
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Clinical Investigation Center, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
| | - Michelle Buettner
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Clinical Investigation Center, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
| | - Kelly Reid
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Clinical Investigation Center, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
| | - Kelly Bergsrud
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Clinical Investigation Center, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
| | - Sara Pracht
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Clinical Investigation Center, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
| | - Andrea Chehadeh
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Clinical Investigation Center, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
| | - Caitlin Feiock
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (R.H.D.); (J.T.S.); (A.J.S.); (H.P.L.); (E.K.); (S.R.); (T.A.D.); (M.L.); (C.F.); (A.R.A.); (M.S.H.); (J.F.M.); (A.B.)
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Clinical Investigation Center, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
| | - M. Gerard O’Sullivan
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA;
| | - Tim Carlson
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA;
| | - Alexandra R. Armstrong
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (R.H.D.); (J.T.S.); (A.J.S.); (H.P.L.); (E.K.); (S.R.); (T.A.D.); (M.L.); (C.F.); (A.R.A.); (M.S.H.); (J.F.M.); (A.B.)
| | - Danielle Meritet
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - Michael S. Henson
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (R.H.D.); (J.T.S.); (A.J.S.); (H.P.L.); (E.K.); (S.R.); (T.A.D.); (M.L.); (C.F.); (A.R.A.); (M.S.H.); (J.F.M.); (A.B.)
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Brenda J. Weigel
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jaime F. Modiano
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (R.H.D.); (J.T.S.); (A.J.S.); (H.P.L.); (E.K.); (S.R.); (T.A.D.); (M.L.); (C.F.); (A.R.A.); (M.S.H.); (J.F.M.); (A.B.)
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Antonella Borgatti
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (R.H.D.); (J.T.S.); (A.J.S.); (H.P.L.); (E.K.); (S.R.); (T.A.D.); (M.L.); (C.F.); (A.R.A.); (M.S.H.); (J.F.M.); (A.B.)
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Clinical Investigation Center, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel A. Vallera
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; (A.L.W.); (K.S.); (M.B.); (K.R.); (K.B.); (S.P.); (A.C.); (M.G.O.); (B.J.W.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Radiation Oncology, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
6
|
Huang M, Park J, Seo J, Ko S, Yang YH, Lee Y, Kim HJ, Lee BS, Lee YS, Ko BJ, Jung ST, Park D, Yoo TH, Kim CH. An epidermal growth factor receptor-targeting immunotoxin based on IgG shows potent antitumor activity against head and neck cancer. FASEB J 2024; 38:e23759. [PMID: 38949635 DOI: 10.1096/fj.202301968r] [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: 09/26/2023] [Revised: 05/18/2024] [Accepted: 06/13/2024] [Indexed: 07/02/2024]
Abstract
The epidermal growth factor receptor (EGFR) is an important target for cancer therapies. Many head and neck cancer (HNC) cells have been reported to overexpress EGFR; therefore, anti-EGFR therapies have been attempted in patients with HNC. However, its clinical efficacy is limited owing to the development of drug resistance. In this study, we developed an EGFR-targeting immunotoxin consisting of a clinically proven anti-EGFR IgG (cetuximab; CTX) and a toxin fragment (LR-LO10) derived from Pseudomonas exotoxin A (PE) using a novel site-specific conjugation technology (peptide-directed photo-crosslinking reaction), as an alternative option. The immunotoxin (CTX-LR-LO10) showed specific binding to EGFR and properties of a typical IgG, such as stability, interactions with receptors of immune cells, and pharmacokinetics, and inhibited protein synthesis via modification of elongation factor-2. Treatment of EGFR-positive HNC cells with the immunotoxin resulted in apoptotic cell death and the inhibition of cell migration and invasion. The efficacy of CTX-LR-LO10 was evaluated in xenograft mouse models, and the immunotoxin exhibited much stronger tumor suppression than CTX or LR-LO10. Transcriptome analyses revealed that the immunotoxins elicited immune responses and altered the expression of genes related to its mechanisms of action. These results support the notion that CTX-LR-LO10 may serve as a new therapeutic agent targeting EGFR-positive cancers.
Collapse
Affiliation(s)
- Mei Huang
- Department of Medical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| | - Jisoo Park
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Jina Seo
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Sanghwan Ko
- Department of Biomedical Sciences, Graduate School, Korea University, Seoul, Republic of Korea
| | - Yoon Hee Yang
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| | - Yeaji Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Hyo Jeong Kim
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Bok-Soon Lee
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Yun Sang Lee
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Byoung Joon Ko
- School of Biopharmaceutical and Medical Sciences, Sungshin Women's University, Seoul, Republic of Korea
| | - Sang Teak Jung
- Department of Biomedical Sciences, Graduate School, Korea University, Seoul, Republic of Korea
- Institute of Human Genetics, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Deachan Park
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
- Advanced College of Bio-convergence Engineering, Ajou University, Suwon, Republic of Korea
| | - Tae Hyeon Yoo
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
- Advanced College of Bio-convergence Engineering, Ajou University, Suwon, Republic of Korea
| | - Chul-Ho Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
7
|
Tang NC, Su JC, Shmidov Y, Kelly G, Deshpande S, Sirohi P, Peterson N, Chilkoti A. Synthetic intrinsically disordered protein fusion tags that enhance protein solubility. Nat Commun 2024; 15:3727. [PMID: 38697982 PMCID: PMC11066018 DOI: 10.1038/s41467-024-47519-7] [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: 08/22/2023] [Accepted: 04/03/2024] [Indexed: 05/05/2024] Open
Abstract
We report the de novo design of small (<20 kDa) and highly soluble synthetic intrinsically disordered proteins (SynIDPs) that confer solubility to a fusion partner with minimal effect on the activity of the fused protein. To identify highly soluble SynIDPs, we create a pooled gene-library utilizing a one-pot gene synthesis technology to create a large library of repetitive genes that encode SynIDPs. We identify three small (<20 kDa) and highly soluble SynIDPs from this gene library that lack secondary structure and have high solvation. Recombinant fusion of these SynIDPs to three known inclusion body forming proteins rescue their soluble expression and do not impede the activity of the fusion partner, thereby eliminating the need for removal of the SynIDP tag. These findings highlight the utility of SynIDPs as solubility tags, as they promote the soluble expression of proteins in E. coli and are small, unstructured proteins that minimally interfere with the biological activity of the fused protein.
Collapse
Affiliation(s)
- Nicholas C Tang
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Jonathan C Su
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Yulia Shmidov
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Garrett Kelly
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Sonal Deshpande
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Parul Sirohi
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Nikhil Peterson
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
8
|
Skorupan N, Peer CJ, Zhang X, Choo-Wosoba H, Ahmad MI, Lee MJ, Rastogi S, Sato N, Yu Y, Pegna GJ, Steinberg SM, Kalsi SS, Cao L, Figg WD, Trepel JB, Pastan I, FitzGerald D, Alewine C. Tofacitinib to prevent anti-drug antibody formation against LMB-100 immunotoxin in patients with advanced mesothelin-expressing cancers. Front Oncol 2024; 14:1386190. [PMID: 38706610 PMCID: PMC11066227 DOI: 10.3389/fonc.2024.1386190] [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: 02/14/2024] [Accepted: 03/22/2024] [Indexed: 05/07/2024] Open
Abstract
Background LMB-100 is a mesothelin (MSLN)-targeting recombinant immunotoxin (iTox) carrying a Pseudomonas exotoxin A payload that has shown promise against solid tumors, however, efficacy is limited by the development of neutralizing anti-drug antibodies (ADAs). Tofacitinib is an oral Janus Kinase (JAK) inhibitor that prevented ADA formation against iTox in preclinical studies. Methods A phase 1 trial testing LMB-100 and tofacitinib in patients with MSLN-expressing cancers (pancreatic adenocarcinoma, n=13; cholangiocarcinoma, n=1; appendiceal carcinoma, n=1; cystadenocarcinoma, n=1) was performed to assess safety and to determine if tofacitinib impacted ADA formation. Participants were treated for up to 3 cycles with LMB-100 as a 30-minute infusion on days 4, 6, and 8 at two dose levels (100 and 140 µg/kg) while oral tofacitinib was administered for the first 10 days of the cycle (10 mg BID). Peripheral blood was collected for analysis of ADA levels, serum cytokines and circulating immune subsets. Results The study was closed early due to occurrence of drug-induced pericarditis in 2 patients. Pericarditis with the combination was not reproducible in a transgenic murine model containing human MSLN. Two of 4 patients receiving all 3 cycles of treatment maintained effective LMB-100 levels, an unusual occurrence. Sustained increases in systemic IL-10 and TNF-α were seen, a phenomenon not observed in prior LMB-100 studies. A decrease in activated T cell subsets and an increase in circulating immunosuppressive myeloid populations occurred. No radiologic decreases in tumor volume were observed. Discussion Further testing of tofacitinib to prevent ADA formation is recommended in applicable non-malignant disease settings. Clinical trial registration https://www.clinicaltrials.gov/study/NCT04034238.
Collapse
Affiliation(s)
- Nebojsa Skorupan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Cody J. Peer
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xianyu Zhang
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Hyoyoung Choo-Wosoba
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mehwish I. Ahmad
- Office of Research Nursing, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Min-Jung Lee
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Shraddha Rastogi
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Nahoko Sato
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yunkai Yu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Guillaume Joe Pegna
- Medical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Shelley S. Kalsi
- Hematology Consult and Graduate Medical Section, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Liang Cao
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - William D. Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jane B. Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - David FitzGerald
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Christine Alewine
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
9
|
Nguyen MQ, Kim DH, Shim HJ, Ta HKK, Vu TL, Nguyen TKO, Lim JC, Choe H. Novel Anti-Mesothelin Nanobodies and Recombinant Immunotoxins with Pseudomonas Exotoxin Catalytic Domain for Cancer Therapeutics. Mol Cells 2023; 46:764-777. [PMID: 38052492 PMCID: PMC10701305 DOI: 10.14348/molcells.2023.0155] [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: 09/01/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 12/07/2023] Open
Abstract
Recombinant immunotoxins (RITs) are fusion proteins consisting of a targeting domain linked to a toxin, offering a highly specific therapeutic strategy for cancer treatment. In this study, we engineered and characterized RITs aimed at mesothelin, a cell surface glycoprotein overexpressed in various malignancies. Through an extensive screening of a large nanobody library, four mesothelin-specific nanobodies were selected and genetically fused to a truncated Pseudomonas exotoxin (PE24B). Various optimizations, including the incorporation of furin cleavage sites, maltose-binding protein tags, and tobacco etch virus protease cleavage sites, were implemented to improve protein expression, solubility, and purification. The RITs were successfully overexpressed in Escherichia coli, achieving high solubility and purity post-purification. In vitro cytotoxicity assays on gastric carcinoma cell lines NCI-N87 and AGS revealed that Meso(Nb2)-PE24B demonstrated the highest cytotoxic efficacy, warranting further characterization. This RIT also displayed selective binding to human and monkey mesothelins but not to mouse mesothelin. The competitive binding assays between different RIT constructs revealed significant alterations in IC50 values, emphasizing the importance of nanobody specificity. Finally, a modification in the endoplasmic reticulum retention signal at the C-terminus further augmented its cytotoxic activity. Our findings offer valuable insights into the design and optimization of RITs, showcasing the potential of Meso(Nb2)-PE24B as a promising therapeutic candidate for targeted cancer treatment.
Collapse
Affiliation(s)
- Minh Quan Nguyen
- Department of Physiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | | | | | - Huynh Kim Khanh Ta
- Department of Physiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Thi Luong Vu
- Department of Physiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Thi Kieu Oanh Nguyen
- Department of Physiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | | | - Han Choe
- Department of Physiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| |
Collapse
|
10
|
Tender GS, Bertozzi CR. Bringing enzymes to the proximity party. RSC Chem Biol 2023; 4:986-1002. [PMID: 38033727 PMCID: PMC10685825 DOI: 10.1039/d3cb00084b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/16/2023] [Indexed: 12/02/2023] Open
Abstract
Enzymes are used to treat a wide variety of human diseases, including lysosomal storage disorders, clotting disorders, and cancers. While enzyme therapeutics catalyze highly specific reactions, they often suffer from a lack of cellular or tissue selectivity. Targeting an enzyme to specific disease-driving cells and tissues can mitigate off-target toxicities and provide novel therapeutic avenues to treat otherwise intractable diseases. Targeted enzymes have been used to treat cancer, in which the enzyme is either carefully selected or engineered to reduce on-target off-tumor toxicity, or to treat lysosomal storage disorders in cell types that are not addressed by standard enzyme replacement therapies. In this review, we discuss the different targeted enzyme modalities and comment on the future of these approaches.
Collapse
Affiliation(s)
- Gabrielle S Tender
- Stanford University, Department of Chemistry and Sarafan ChEM-H Stanford CA 94305 USA
| | - Carolyn R Bertozzi
- Stanford University, Department of Chemistry and Sarafan ChEM-H Stanford CA 94305 USA
- Howard Hughes Medical Institute Stanford CA 94305 USA
| |
Collapse
|
11
|
Fischer A, Masilamani AP, Schultze-Seemann S, Wolf I, Gratzke C, Fuchs H, Wolf P. Synergistic Cytotoxicity of a Toxin Targeting the Epidermal Growth Factor Receptor and the Glycosylated Triterpenoid SO1861 in Prostate Cancer. J Cancer 2023; 14:3039-3049. [PMID: 37859824 PMCID: PMC10583583 DOI: 10.7150/jca.85691] [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: 04/27/2023] [Accepted: 08/01/2023] [Indexed: 10/21/2023] Open
Abstract
Treatment of advanced prostate cancer lacks specificity and curative intent. Therefore, the need for new targeted therapeutic approaches is high. In the present study, we generated the new targeted toxin EGF-PE24mutΔREDLK binding to the epidermal growth factor receptor (EGFR) on the surface of prostate cancer cells. It consists of the human epidermal growth factor (EGF) as binding domain and a de-immunized variant of Pseudomonas Exotoxin A (PE), called PE24mutΔREDLK, as toxin domain. The toxin domain contains a deletion of the C-terminal KDEL-like motif REDLK to prevent its transport from sorting endosomes via the KDEL receptor mediated pathway into the cytosol, where it can inhibit cellular protein biosynthesis and induce apoptosis. Indeed, REDLK deletion resulted in a strong decrease in cytotoxicity of the targeted toxin in prostate cancer cells compared to the parental targeted toxin EGF-PE24mut. However, addition of the plant glycosylated triterpenoid SO1861, which is known to mediate the release of biomolecules from endolysosomal compartments into the cytosol, resulted in an up to almost 7,000-fold enhanced synergistic cytotoxicity. Moreover, combination of PE24mutΔREDLK with SO1861 led to a cytotoxicity that was even 16- to 300-fold enhanced compared to that of EGF-PE24mut. Endolysosomal entrapment of the non-toxic targeted toxin EGF-PE24mutΔREDLK followed by activation through enhanced endosomal escape therefore represents a new promising approach for the future treatment of advanced prostate cancer with high efficacy and diminished side effects.
Collapse
Affiliation(s)
- Alexandra Fischer
- Department of Urology, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anie Priscilla Masilamani
- Department of Urology, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Susanne Schultze-Seemann
- Department of Urology, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Isis Wolf
- Department of Urology, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty for Biology, University of Freiburg, Freiburg, Germany
| | - Christian Gratzke
- Department of Urology, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hendrik Fuchs
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry; Berlin, Germany
| | - Philipp Wolf
- Department of Urology, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
12
|
Liu X, Tan Q, Wen J, Wang X, Yang G, Li Y, Lu M, Ye W, Si A, Ma S, Ding T, Sun L, Liu F, Zhang M, Jiang T, Gao W. Improving the cytotoxicity of immunotoxins by reducing the affinity of the antibody in acidic pH. J Transl Med 2023; 21:572. [PMID: 37626430 PMCID: PMC10463491 DOI: 10.1186/s12967-023-04210-7] [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: 12/24/2022] [Accepted: 05/19/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Immunotoxins are antibody-toxin conjugates that bind to surface antigens and exert effective cytotoxic activity after internalization into tumor cells. Immunotoxins exhibit effective cytotoxicity and have been approved by the FDA to treat multiple hematological malignancies, such as hairy cell leukemia and cutaneous T-cell lymphoma. However, most of the internalized immunotoxin is degraded in lysosomes, and only approximately 5% of free toxin escapes into the cytosol to exert cytotoxicity. Many studies have improved immunotoxins by engineering the toxin fragment to reduce immunogenicity or increase stability, but how the antibody fragment contributes to the activity of immunotoxins has not been well demonstrated. METHODS In the current study, we used 32A9 and 42A1, two anti-GPC3 antibodies with similar antigen-binding capabilities and internalization rates, to construct scFv-mPE24 immunotoxins and evaluated their in vitro and in vivo antitumor activities. Next, the antigen-binding capacity, trafficking, intracellular protein stability and release of free toxin of 32A9 scFv-mPE24 and 42A1 scFv-mPE24 were compared to elucidate their different antitumor activities. Furthermore, we used a lysosome inhibitor to evaluate the degradation behavior of 32A9 scFv-mPE24 and 42A1 scFv-mPE24. Finally, the antigen-binding patterns of 32A9 and 42A1 were compared under neutral and acidic pH conditions. RESULTS Although 32A9 and 42A1 had similar antigen binding capacities and internalization rates, 32A9 scFv-mPE24 had superior antitumor activity compared to 42A1 scFv-mPE24. We found that 32A9 scFv-mPE24 exhibited faster degradation and drove efficient free toxin release compared to 42A1 scFv-mPE24. These phenomena were determined by the different degradation behaviors of 32A9 scFv-mPE24 and 42A1 scFv-mPE24 in lysosomes. Moreover, 32A9 was sensitive to the low-pH environment, which made the 32A9 conjugate easily lose antigen binding and undergo degradation in lysosomes, and the free toxin was then efficiently produced to exert cytotoxicity, whereas 42A1 was resistant to the acidic environment, which kept the 42A1 conjugate relatively stable in lysosomes and delayed the release of free toxin. CONCLUSIONS These results showed that a low pH-sensitive antibody-based immunotoxin degraded faster in lysosomes, caused effective free toxin release, and led to improved cytotoxicity compared to an immunotoxin based on a normal antibody. Our findings suggested that a low pH-sensitive antibody might have an advantage in the design of immunotoxins and other lysosomal degradation-dependent antibody conjugate drugs.
Collapse
Affiliation(s)
- Xiaoyu Liu
- School of Basic Medical Sciences and Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Qingqing Tan
- Department of Gynecology Oncology, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Jiaqi Wen
- School of Basic Medical Sciences and Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Xufei Wang
- School of Basic Medical Sciences and Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Gang Yang
- School of Basic Medical Sciences and Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Yuxiao Li
- Department of Endocrinology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Ming Lu
- School of Basic Medical Sciences and Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Wei Ye
- School of Basic Medical Sciences and Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Anfeng Si
- Department of Surgical Oncology, Jinling Hospital, Medical School of Nanjing University, 34 Yanggongjing Road, Nanjing, 210000, Jiangsu, People's Republic of China
| | - Sujuan Ma
- School of Basic Medical Sciences and Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Tong Ding
- School of Basic Medical Sciences and Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Luan Sun
- School of Basic Medical Sciences and Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Fang Liu
- School of Basic Medical Sciences and Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Mei Zhang
- Department of Endocrinology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Tao Jiang
- Department of Surgical Oncology, Jinling Hospital, Medical School of Nanjing University, 34 Yanggongjing Road, Nanjing, 210000, Jiangsu, People's Republic of China.
| | - Wei Gao
- School of Basic Medical Sciences and Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Nanjing, 211166, Jiangsu, People's Republic of China.
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China.
| |
Collapse
|
13
|
Masilamani AP, Huber N, Nagl C, Dettmer-Monaco V, Monaco G, Wolf I, Schultze-Seemann S, Taromi S, Gratzke C, Fuchs H, Wolf P. Enhanced cytotoxicity of a Pseudomonas Exotoxin A based immunotoxin against prostate cancer by addition of the endosomal escape enhancer SO1861. Front Pharmacol 2023; 14:1211824. [PMID: 37484018 PMCID: PMC10358361 DOI: 10.3389/fphar.2023.1211824] [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: 04/25/2023] [Accepted: 06/27/2023] [Indexed: 07/25/2023] Open
Abstract
Immunotoxins consist of an antibody or antibody fragment that binds to a specific cell surface structure and a cytotoxic domain that kills the cell after cytosolic uptake. Pseudomonas Exotoxin A (PE) based immunotoxins directed against a variety of tumor entities have successfully entered the clinic. PE possesses a KDEL-like motif (REDLK) that enables the toxin to travel from sorting endosomes via the KDEL-receptor pathway to the endoplasmic reticulum (ER), from where it is transported into the cytosol. There, it ADP-ribosylates the eukaryotic elongation factor 2, resulting in ribosome inhibition and finally apoptosis. One major problem of immunotoxins is their lysosomal degradation causing the need for much more immunotoxin molecules than finally required for induction of cell death. The resulting dose limitations and substantially increased side effects require new strategies to achieve improved cytosolic uptake. Here we generated an immunotoxin consisting of a humanized single chain variable fragment (scFv) targeting the prostate specific membrane antigen (PSMA) and the de-immunized PE variant PE24mut. This immunotoxin, hD7-1(VL-VH)-PE24mut, showed high and specific cytotoxicity in PSMA-expressing prostate cancer cells. We deleted the REDLK sequence to prevent transport to the ER and achieve endosomal entrapment. The cytotoxicity of this immunotoxin, hD7-1(VL-VH)-PE24mutΔREDLK, was greatly reduced. To restore activity, we added the endosomal escape enhancer SO1861 and observed an up to 190,000-fold enhanced cytotoxicity corresponding to a 57-fold enhancement compared to the initial immunotoxin with the REDLK sequence. A biodistribution study with different routes of administration clearly showed that the subcutaneous injection of hD7-1(VL-VH)-PE24mutΔREDLK in mice resulted in the highest tumor uptake. Treatment of mice bearing prostate tumors with a combination of hD7-1(VL-VH)-PE24mutΔREDLK plus SO1861 resulted in inhibition of tumor growth and enhanced overall survival compared to the monotherapies. The endosomal entrapment of non-toxic anti-PSMA immunotoxins followed by enhanced endosomal escape by SO1861 provides new therapeutic options in the future management of prostate cancer.
Collapse
Affiliation(s)
- Anie P. Masilamani
- Department of Urology, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nathalie Huber
- Department of Urology, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Constanze Nagl
- Department of Urology, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Viviane Dettmer-Monaco
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Transfusion Medicine and Gene Therapy, Medical Center—University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| | - Gianni Monaco
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Transfusion Medicine and Gene Therapy, Medical Center—University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Institute of Neuropathology, Medical Center—University of Freiburg, Freiburg, Germany
| | - Isis Wolf
- Department of Urology, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty for Biology, University of Freiburg, Freiburg, Germany
| | - Susanne Schultze-Seemann
- Department of Urology, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sanaz Taromi
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Medicine I, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medical and Life Sciences, University Furtwangen, VS-Schwenningen, Germany
| | - Christian Gratzke
- Department of Urology, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hendrik Fuchs
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp Wolf
- Department of Urology, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
14
|
Chen Z, Wang X, Chen X, Huang J, Wang C, Wang J, Wang Z. Accelerating therapeutic protein design with computational approaches toward the clinical stage. Comput Struct Biotechnol J 2023; 21:2909-2926. [PMID: 38213894 PMCID: PMC10781723 DOI: 10.1016/j.csbj.2023.04.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/11/2023] [Accepted: 04/27/2023] [Indexed: 01/13/2024] Open
Abstract
Therapeutic protein, represented by antibodies, is of increasing interest in human medicine. However, clinical translation of therapeutic protein is still largely hindered by different aspects of developability, including affinity and selectivity, stability and aggregation prevention, solubility and viscosity reduction, and deimmunization. Conventional optimization of the developability with widely used methods, like display technologies and library screening approaches, is a time and cost-intensive endeavor, and the efficiency in finding suitable solutions is still not enough to meet clinical needs. In recent years, the accelerated advancement of computational methodologies has ushered in a transformative era in the field of therapeutic protein design. Owing to their remarkable capabilities in feature extraction and modeling, the integration of cutting-edge computational strategies with conventional techniques presents a promising avenue to accelerate the progression of therapeutic protein design and optimization toward clinical implementation. Here, we compared the differences between therapeutic protein and small molecules in developability and provided an overview of the computational approaches applicable to the design or optimization of therapeutic protein in several developability issues.
Collapse
Affiliation(s)
- Zhidong Chen
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Xinpei Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Xu Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Juyang Huang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Chenglin Wang
- Shenzhen Qiyu Biotechnology Co., Ltd, Shenzhen 518107, China
| | - Junqing Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Zhe Wang
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| |
Collapse
|
15
|
Rhee JH, Khim K, Puth S, Choi Y, Lee SE. Deimmunization of flagellin adjuvant for clinical application. Curr Opin Virol 2023; 60:101330. [PMID: 37084463 DOI: 10.1016/j.coviro.2023.101330] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 04/23/2023]
Abstract
Flagellin is the cognate ligand for host pattern recognition receptors, toll-like receptor 5 (TLR5) in the cell surface, and NAIP5/NLRC4 inflammasome in the cytosol. TLR5-binding domain is located in D1 domain, where crucial amino acid sequences are conserved among diverse bacteria. The highly conserved C-terminal 35 amino acids of flagellin were proved to be responsible for the inflammasome activation by binding to NAIP5. D2/D3 domains, located in the central region and exposed to the outside surface of flagellar filament, are heterogeneous across bacterial species and highly immunogenic. Taking advantage of TLR5- and NLRC4-stimulating activities, flagellin has been actively developed as a vaccine adjuvant and immunotherapeutic. Because of its immunogenicity, there exist worries concerning diminished efficacy and possible reactogenicity after repeated administration. Deimmunization of flagellin derivatives while preserving the TLR5/NLRC4-mediated immunomodulatory activity should be the most reasonable option for clinical application. This review describes strategies and current achievements in flagellin deimmunization.
Collapse
Affiliation(s)
- Joon Haeng Rhee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea; Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea; Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea.
| | - Koemchhoy Khim
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea; Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Sao Puth
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea; Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Yoonjoo Choi
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea; Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Shee Eun Lee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea; Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
| |
Collapse
|
16
|
Xu T, Schulga A, Konovalova E, Rinne SS, Zhang H, Vorontsova O, Orlova A, Deyev SM, Tolmachev V, Vorobyeva A. Feasibility of Co-Targeting HER3 and EpCAM Using Seribantumab and DARPin-Toxin Fusion in a Pancreatic Cancer Xenograft Model. Int J Mol Sci 2023; 24:ijms24032838. [PMID: 36769161 PMCID: PMC9917732 DOI: 10.3390/ijms24032838] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most aggressive malignancies. A combination of targeted therapies could increase the therapeutic efficacy in tumors with heterogeneous target expression. Overexpression of the human epidermal growth factor receptor type 3 (HER3) and the epithelial cell adhesion molecule (EpCAM) in up to 40% and 30% of PCs, respectively, is associated with poor prognosis and highlights the relevance of these targets. Designed ankyrin repeat protein (DARPin) Ec1 fused with the low immunogenic bacterial toxin LoPE provides specific and potent cytotoxicity against EpCAM-expressing cancer cells. Here, we investigated whether the co-targeting of HER3 using the monoclonal antibody seribantumab (MM-121) and of EpCAM using Ec1-LoPE would improve the therapeutic efficacy in comparison to the individual agents. Radiolabeled 99mTc(CO)3-Ec1-LoPE showed specific binding with rapid internalization in EpCAM-expressing PC cells. MM-121 did not interfere with the binding of Ec1-LoPE to EpCAM. Evaluation of cytotoxicity indicated synergism between Ec1-LoPE and MM-121 in vitro. An experimental therapy study using Ec1-LoPE and MM-121 in mice bearing EpCAM- and HER3-expressing BxPC3 xenografts demonstrated the feasibility of the therapy. Further development of the co-targeting approach using HER3 and EpCAM could therefore be justified.
Collapse
Affiliation(s)
- Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Alexey Schulga
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk 634050, Russia
- Molecular Immunology Laboratory, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Elena Konovalova
- Molecular Immunology Laboratory, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Hongchao Zhang
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Olga Vorontsova
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk 634050, Russia
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, 751 23 Uppsala, Sweden
| | - Sergey M. Deyev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk 634050, Russia
- Molecular Immunology Laboratory, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Bio-Nanophotonic Laboratory, Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University ‘MEPhI’, Moscow 115409, Russia
- Center of Biomedical Engineering, Sechenov University, Moscow 119991, Russia
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk 634050, Russia
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
- Correspondence:
| |
Collapse
|
17
|
Nishio T, Koyama Y, Fuji H, Ishizuka K, Iwaisako K, Taura K, Hatano E, Brenner DA, Kisseleva T. The Role of Mesothelin in Activation of Portal Fibroblasts in Cholestatic Liver Injury. BIOLOGY 2022; 11:1589. [PMID: 36358290 PMCID: PMC9687690 DOI: 10.3390/biology11111589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/18/2022] [Accepted: 10/27/2022] [Indexed: 11/05/2022]
Abstract
Fibrosis is a common consequence of abnormal wound healing, which is characterized by infiltration of myofibroblasts and formation of fibrous scar. In liver fibrosis, activated Hepatic Stellate Cells (aHSCs) and activated Portal Fibroblasts (aPFs) are the major contributors to the origin of hepatic myofibroblasts. aPFs are significantly involved in the pathogenesis of cholestatic fibrosis, suggesting that aPFs may be a primary target for anti-fibrotic therapy in cholestatic injury. aPFs are distinguishable from aHSCs by specific markers including mesothelin (Msln), Mucin 16 (Muc16), and Thymus cell antigen 1 (Thy1, CD90) as well as fibulin 2, elastin, Gremlin 1, ecto-ATPase nucleoside triphosphate diphosphohydrolase 2. Msln plays a critical role in activation of PFs, via formation of Msln-Muc16-Thy1 complex that regulates TGFβ1/TGFβRI-mediated fibrogenic signaling. The opposing pro- and anti-fibrogenic effects of Msln and Thy1 are key components of the TGFβ1-induced activation pathway in aPFs. In addition, aPFs and activated lung and kidney fibroblasts share similarities across different organs with expression of common markers and activation cascade including Msln-Thy1 interaction. Here, we summarize the potential function of Msln in activation of PFs and development of cholestatic fibrosis, offering a novel perspective for anti-fibrotic therapy targeting Msln.
Collapse
Affiliation(s)
- Takahiro Nishio
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
- Department of Surgery, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yukinori Koyama
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hiroaki Fuji
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
- Department of Surgery, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
| | - Kei Ishizuka
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
- Department of Surgery, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
| | - Keiko Iwaisako
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, 1-3 Tataramiyakodani, Kyotanabe 610-0394, Japan
| | - Kojiro Taura
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
- Department of Gastroenterological Surgery and Oncology, Kitano Hospital Medical Research Institute, 2-4-20 Ogimachi, Kita-ku, Osaka 530-8480, Japan
| | - Etsuro Hatano
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - David A. Brenner
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
| | - Tatiana Kisseleva
- Department of Surgery, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
| |
Collapse
|
18
|
Shen X, Lin Q, Liang Z, Wang J, Yang X, Liang Y, Liang H, Pan H, Yang J, Zhu Y, Li M, Xiang W, Zhu H. Reduction of Pre-Existing Adaptive Immune Responses Against SaCas9 in Humans Using Epitope Mapping and Identification. CRISPR J 2022; 5:445-456. [PMID: 35686980 DOI: 10.1089/crispr.2021.0142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The CRISPR-Cas9 system is increasingly being used as a gene editing therapeutic technique in complex diseases but concerns remain regarding the clinical risks of Cas9 immunogenicity. In this study, we detected antibodies against Staphylococcus aureus Cas9 (SaCas9) and anti-SaCas9 T cells in 4.8% and 70% of Chinese donors, respectively. We predicted 135 SaCas9-derived B cell epitopes and 50 SaCas9-derived CD8+ T cell epitopes for HLA-A*24:02, HLA-A*11:01, and HLA-A*02:01. We identified R338 as an immunodominant SaCas9 B cell epitope and SaCas9_200-208 as an immunodominant CD8+ T cell epitope for the three human leukocyte antigen allotypes through immunological assays using sera positive for SaCas9-specific antibodies and peripheral blood mononuclear cells positive for SaCas9-reactive T cells, respectively. We also demonstrated that an SaCas9 variant bearing an R338G substitution reduces B cell immunogenicity and retains its gene-editing function. Our study highlights the immunological risks of the CRISPR-Cas9 system and provides a solution to mitigate pre-existing adaptive immune responses against Cas9 in humans.
Collapse
Affiliation(s)
- Xiaoting Shen
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Qinru Lin
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Zhiming Liang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Jing Wang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Xinyi Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Yue Liang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Huitong Liang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Hanyu Pan
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Jinlong Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Yuqi Zhu
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Min Li
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Weirong Xiang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Huanzhang Zhu
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Xu T, Liu Y, Schulga A, Konovalova E, Deyev S, Tolmachev V, Vorobyeva A. Epithelial cell adhesion molecule‑targeting designed ankyrin repeat protein‑toxin fusion Ec1‑LoPE exhibits potent cytotoxic action in prostate cancer cells. Oncol Rep 2022; 47:94. [PMID: 35315504 PMCID: PMC8968790 DOI: 10.3892/or.2022.8305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
Targeted anticancer therapeutics offer the advantage of reducing cytotoxic side effects to normal cells by directing the cytotoxic payload selectively to cancer cells. Designed ankyrin repeat proteins (DARPins) are promising non-immunoglobulin-based scaffold proteins for payload delivery to cancer-associated molecular targets. Epithelial cell adhesion molecule (EpCAM) is overexpressed in 40–60% of prostate cancers (PCs) and is associated with metastasis, increased risk of PC recurrence and resistance to treatment. Here, we investigated the use of DARPin Ec1 for targeted delivery of Pseudomonas exotoxin A variant (LoPE) with low immunogenicity and low non-specific toxicity to EpCAM-expressing prostate cancer cells. Ec1-LoPE fusion protein was radiolabeled with tricarbonyl technetium-99m and its binding specificity, binding kinetics, cellular processing, internalization and cytotoxicity were evaluated in PC-3 and DU145 cell lines. Ec1-LoPE showed EpCAM-specific binding to EpCAM-expressing prostate cancer cells. Rapid internalization mediated potent cytotoxic effect with picomolar IC50 values in both studied cell lines. Taken together, these data support further evaluation of Ec1-LoPE in a therapeutic setting in a prostate cancer model in vivo.
Collapse
Affiliation(s)
- Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Alexey Schulga
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Elena Konovalova
- Molecular Immunology Laboratory, Shemyakin‑Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Sergey Deyev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| |
Collapse
|
20
|
Shipunova VO, Deyev SM. Artificial Scaffold Polypeptides As an Efficient Tool for the Targeted Delivery of Nanostructures In Vitro and In Vivo. Acta Naturae 2022; 14:54-72. [PMID: 35441046 PMCID: PMC9013437 DOI: 10.32607/actanaturae.11545] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/20/2021] [Indexed: 12/22/2022] Open
Abstract
The use of traditional tools for the targeted delivery of nanostructures, such as antibodies, transferrin, lectins, or aptamers, often leads to an entire range of undesirable effects. The large size of antibodies often does not allow one to reach the required number of molecules on the surface of nanostructures during modification, and the constant domains of heavy chains, due to their effector functions, can induce phagocytosis. In the recent two decades, targeted polypeptide scaffold molecules of a non-immunoglobulin nature, antibody mimetics, have emerged as much more effective targeting tools. They are small in size (3-20 kDa), possess high affinity (from subnano- to femtomolar binding constants), low immunogenicity, and exceptional thermodynamic stability. These molecules can be effectively produced in bacterial cells, and, using genetic engineering manipulations, it is possible to create multispecific fusion proteins for the targeting of nanoparticles to cells with a given molecular portrait, which makes scaffold polypeptides an optimal tool for theranostics.
Collapse
Affiliation(s)
- V. O. Shipunova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russia
| | - S. M. Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russia
| |
Collapse
|
21
|
Havaei SM, Aucoin MG, Jahanian-Najafabadi A. Pseudomonas Exotoxin-Based Immunotoxins: Over Three Decades of Efforts on Targeting Cancer Cells With the Toxin. Front Oncol 2021; 11:781800. [PMID: 34976821 PMCID: PMC8716853 DOI: 10.3389/fonc.2021.781800] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/16/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer is one of the prominent causes of death worldwide. Despite the existence of various modalities for cancer treatment, many types of cancer remain uncured or develop resistance to therapeutic strategies. Furthermore, almost all chemotherapeutics cause a range of side effects because they affect normal cells in addition to malignant cells. Therefore, the development of novel therapeutic agents that are targeted specifically toward cancer cells is indispensable. Immunotoxins (ITs) are a class of tumor cell-targeted fusion proteins consisting of both a targeting moiety and a toxic moiety. The targeting moiety is usually an antibody/antibody fragment or a ligand of the immune system that can bind an antigen or receptor that is only expressed or overexpressed by cancer cells but not normal cells. The toxic moiety is usually a protein toxin (or derivative) of animal, plant, insect, or bacterial origin. To date, three ITs have gained Food and Drug Administration (FDA) approval for human use, including denileukin diftitox (FDA approval: 1999), tagraxofusp (FDA approval: 2018), and moxetumomab pasudotox (FDA approval: 2018). All of these ITs take advantage of bacterial protein toxins. The toxic moiety of the first two ITs is a truncated form of diphtheria toxin, and the third is a derivative of Pseudomonas exotoxin (PE). There is a growing list of ITs using PE, or its derivatives, being evaluated preclinically or clinically. Here, we will review these ITs to highlight the advances in PE-based anticancer strategies, as well as review the targeting moieties that are used to reduce the non-specific destruction of non-cancerous cells. Although we tried to be as comprehensive as possible, we have limited our review to those ITs that have proceeded to clinical trials and are still under active clinical evaluation.
Collapse
Affiliation(s)
- Seyed Mehdi Havaei
- 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
|
22
|
Fuji H, Miller G, Nishio T, Koyama Y, Lam K, Zhang V, Loomba R, Brenner D, Kisseleva T. The role of Mesothelin signaling in Portal Fibroblasts in the pathogenesis of cholestatic liver fibrosis. Front Mol Biosci 2021; 8:790032. [PMID: 34966784 PMCID: PMC8710774 DOI: 10.3389/fmolb.2021.790032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/15/2021] [Indexed: 01/18/2023] Open
Abstract
Liver fibrosis develops in response to chronic toxic or cholestatic injury, and is characterized by apoptosis of damaged hepatocytes, development of inflammatory responses, and activation of Collagen Type I producing myofibroblasts that make liver fibrotic. Two major cell types, Hepatic Stellate Cells (HSCs) and Portal Fibroblasts (PFs) are the major source of hepatic myofibroblasts. Hepatotoxic liver injury activates Hepatic Stellate Cells (aHSCs) to become myofibroblasts, while cholestatic liver injury activates both aHSCs and Portal Fibroblasts (aPFs). aPFs comprise the major population of myofibroblasts at the onset of cholestatic injury, while aHSCs are increasingly activated with fibrosis progression. Here we summarize our current understanding of the role of aPFs in the pathogenesis of cholestatic fibrosis, their unique features, and outline the potential mechanism of targeting aPFs in fibrotic liver.
Collapse
Affiliation(s)
- Hiroaki Fuji
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
| | - Grant Miller
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
| | - Takahiro Nishio
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yukinori Koyama
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kevin Lam
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
| | - Vivian Zhang
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
| | - Rohit Loomba
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - David Brenner
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Tatiana Kisseleva
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
23
|
Engineering of Cytolethal Distending Toxin B by Its Reducing Immunogenicity and Maintaining Stability as a New Drug Candidate for Tumor Therapy; an In Silico Study. Toxins (Basel) 2021; 13:toxins13110785. [PMID: 34822569 PMCID: PMC8624547 DOI: 10.3390/toxins13110785] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 10/30/2021] [Accepted: 11/01/2021] [Indexed: 12/25/2022] Open
Abstract
The cytolethal distending toxin (CDT), Haemophilus ducreyi, is one of the bacterial toxins that have recently been considered for targeted therapies, especially in cancer therapies. CDT is an A-B2 exotoxin. Its catalytic subunit (CdtB) is capable of inducing DNA double strand breaks, cell cycle arrest and apoptosis in host eukaryotic cells. The sequence alignment indicates that the CdtB is structurally homologyr to phosphatases and deoxyribonucleases I (DNase I). Recently, it has been found that CdtB toxicity is mainly related to its nuclease activity. The immunogenicity of CDT can reduce its effectiveness in targeted therapies. However, the toxin can be very useful if its immunogenicity is significantly reduced. Detecting hotspot ectopic residues by computational servers and then mutating them to eliminate B-cell epitopes is a promising approach to reduce the immunogenicity of foreign protein-based therapeutics. By the mentioned method, in this study, we try to reduce the immunogenicity of the CdtB- protein sequence. This study initially screened residue of the CdtB is B-cell epitopes both linearly and conformationally. By overlapping the B-cell epitopes with the excluded conserve residues, and active and enzymatic sites, four residues were allowed to be mutated. There were two mutein options that show reduced antigenicity probability. Option one was N19F, G74I, and S161F with a VaxiJen score of 0.45 and the immune epitope database (IEDB) score of 1.80, and option two was N19F, G74I, and S161W with a VaxiJen score of 0.45 and IEDB score of 1.88. The 3D structure of the proposed sequences was evaluated and refined. The structural stability of native and mutant proteins was accessed through molecular dynamic simulation. The results showed that the mutations in the mutants caused no considerable changes in their structural stability. However, mutant 1 reveals more thermodynamic stability during the simulation. The applied approaches in this study can be used as rough guidelines for finding hot spot immunogen regions in the therapeutic proteins. Our results provide a new version of CdtB that, due to reduced immunogenicity and increased stability, can be used in toxin-based drugs such as immunotoxins.
Collapse
|
24
|
Immunotoxin IHP25-BT with low immunogenicity and off-target toxicity inhibits the growth and metastasis of trastuzumab-resistant tumor cells. Int J Pharm 2021; 608:121081. [PMID: 34506924 DOI: 10.1016/j.ijpharm.2021.121081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/22/2021] [Accepted: 09/04/2021] [Indexed: 12/30/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) is overexpressed in some breast and gastric cancer patients. As the first HER2-targeteed therpeutic antibody, trastuzumab could significantly improve the prognosis of HER2-positive cancer patients. However, even responding patients inevitably get worse due to acquired resistance to trastuzumab after a period of treatment. Many HER2-targeted antibody drugs used wild-type tumor cells to conduct their corresponding preclinical experiments in vitro and in vivo. However, it is impossible to determine whether these newly developed drugs have antitumor effective to trastuzumab-resistant tumor cells. In the study, two trastuzumab-resistant HER2-positive tumor cell populations NCI-N87-TR and BT474-TR were generated. Then, we examined the anti-tumor effects of newly constructed immunotoxins with low immunogenicity and off-target toxicity based on the trastuzumab-resistant tumor cells both in vitro and in vivo. Results demonstrated that the immunotoxin IHP25-BT could not only effectively inhibit tumor growth but also inhibit liver metastasis of tumor cells in a mouse xenograft model. Furthermore, tumor tissue transcriptome sequencing was performed to clarify the potential mechanisms of inhibiting tumor cell distant metastasis by immunotoxin. In conclusion, this work describes a series of attractive therapeutic immunotoxins, the low immunogenicity and off-target toxicity making them promising for trastuzumab-resistant cancer therapy.
Collapse
|
25
|
Xu T, Vorobyeva A, Schulga A, Konovalova E, Vorontsova O, Ding H, Gräslund T, Tashireva LA, Orlova A, Tolmachev V, Deyev SM. Imaging-Guided Therapy Simultaneously Targeting HER2 and EpCAM with Trastuzumab and EpCAM-Directed Toxin Provides Additive Effect in Ovarian Cancer Model. Cancers (Basel) 2021; 13:3939. [PMID: 34439094 PMCID: PMC8393281 DOI: 10.3390/cancers13163939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/23/2021] [Accepted: 08/02/2021] [Indexed: 12/29/2022] Open
Abstract
Efficient treatment of disseminated ovarian cancer (OC) is challenging due to its heterogeneity and chemoresistance. Overexpression of human epidermal growth factor receptor 2 (HER2) and epithelial cell adhesion molecule (EpCAM) in approx. 30% and 70% of ovarian cancers, respectively, allows for co-targeted treatment. The clinical efficacy of the monoclonal antibody trastuzumab in patients with HER2-positive breast, gastric and gastroesophageal cancers makes it readily available as the HER2-targeting component. As the EpCAM-targeting component, we investigated the designed ankyrin repeat protein (DARPin) Ec1 fused to a truncated variant of Pseudomonas exotoxin A with reduced immunogenicity and low general toxicity (LoPE). Ec1-LoPE was radiolabeled, evaluated in ovarian cancer cells in vitro and its biodistribution and tumor-targeting properties were studied in vivo. The therapeutic efficacy of Ec1-LoPE alone and in combination with trastuzumab was studied in mice bearing EpCAM- and HER2-expressing SKOV3 xenografts. SPECT/CT imaging enabled visualization of EpCAM and HER2 expression in the tumors. Co-treatment using Ec1-LoPE and trastuzumab was more effective at reducing tumor growth and prolonged the median survival of mice compared with mice in the control and monotherapy groups. Repeated administration of Ec1-LoPE was well tolerated without signs of hepatic or kidney toxicity. Co-treatment with trastuzumab and Ec1-LoPE might be a potential therapeutic strategy for HER2- and EpCAM-positive OC.
Collapse
Affiliation(s)
- Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (O.V.)
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (O.V.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (A.S.); (A.O.); (S.M.D.)
| | - Alexey Schulga
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (A.S.); (A.O.); (S.M.D.)
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
| | - Elena Konovalova
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
| | - Olga Vorontsova
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (O.V.)
| | - Haozhong Ding
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden; (H.D.); (T.G.)
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden; (H.D.); (T.G.)
| | - Liubov A. Tashireva
- Cancer Research Institute, Tomsk National Research Medical Center Russian Academy of Sciences, 634009 Tomsk, Russia;
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (A.S.); (A.O.); (S.M.D.)
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, 751 23 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (O.V.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (A.S.); (A.O.); (S.M.D.)
| | - Sergey M. Deyev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (A.S.); (A.O.); (S.M.D.)
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
- Bio-Nanophotonic Lab, Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University ‘MEPhI’, 115409 Moscow, Russia
- Center of Biomedical Engineering, Sechenov University, 119991 Moscow, Russia
| |
Collapse
|
26
|
Immunotherapy-based targeting of MSLN + activated portal fibroblasts is a strategy for treatment of cholestatic liver fibrosis. Proc Natl Acad Sci U S A 2021; 118:2101270118. [PMID: 34253615 DOI: 10.1073/pnas.2101270118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
We investigated the role of mesothelin (Msln) and thymocyte differentiation antigen 1 (Thy1) in the activation of fibroblasts across multiple organs and demonstrated that Msln-/- mice are protected from cholestatic fibrosis caused by Mdr2 (multidrug resistance gene 2) deficiency, bleomycin-induced lung fibrosis, and UUO (unilateral urinary obstruction)-induced kidney fibrosis. On the contrary, Thy1-/- mice are more susceptible to fibrosis, suggesting that a Msln-Thy1 signaling complex is critical for tissue fibroblast activation. A similar mechanism was observed in human activated portal fibroblasts (aPFs). Targeting of human MSLN+ aPFs with two anti-MSLN immunotoxins killed fibroblasts engineered to express human mesothelin and reduced collagen deposition in livers of bile duct ligation (BDL)-injured mice. We provide evidence that antimesothelin-based therapy may be a strategy for treatment of parenchymal organ fibrosis.
Collapse
|
27
|
Dellas N, Liu J, Botham RC, Huisman GW. Adapting protein sequences for optimized therapeutic efficacy. Curr Opin Chem Biol 2021; 64:38-47. [PMID: 33933937 DOI: 10.1016/j.cbpa.2021.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 12/24/2022]
Abstract
Therapeutic proteins alleviate disease pathology by supplementing missing or defective native proteins, sequestering superfluous proteins, or by acting through designed non-natural mechanisms. Although therapeutic proteins often have the same amino acid sequence as their native counterpart, their maturation paths from expression to the site of physiological activity are inherently different, and optimizing protein sequences for properties that 100s of millions of years of evolution did not need to address presents an opportunity to develop better biological treatments. Because therapeutic proteins are inherently non-natural entities, optimization for their desired function should be considered analogous to that of small molecule drug candidates, which are optimized through expansive combinatorial variation by the medicinal chemist. Here, we review recent successes and challenges of protein engineering for optimized therapeutic efficacy.
Collapse
Affiliation(s)
- Nikki Dellas
- Codexis Inc., 200 Penobscot Dr, Redwood City, CA, 94063, USA.
| | - Joyce Liu
- Codexis Inc., 200 Penobscot Dr, Redwood City, CA, 94063, USA
| | - Rachel C Botham
- Codexis Inc., 200 Penobscot Dr, Redwood City, CA, 94063, USA
| | - Gjalt W Huisman
- Codexis Inc., 200 Penobscot Dr, Redwood City, CA, 94063, USA
| |
Collapse
|
28
|
Dashtiahangar M, Rahbarnia L, Farajnia S, Salmaninejad A, Shabgah AG, Ghasemali S. Anti-cancer Immunotoxins, Challenges, and Approaches. Curr Pharm Des 2021; 27:932-941. [PMID: 33023437 DOI: 10.2174/1381612826666201006155346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/07/2020] [Indexed: 11/22/2022]
Abstract
The development of recombinant immunotoxins (RITs) as a novel therapeutic strategy has made a revolution in the treatment of cancer. RITs result from the fusion of antibodies to toxin proteins for targeting and eliminating cancerous cells by inhibiting protein synthesis. Despite indisputable outcomes of RITs regarding inhibition of multiple cancer types, high immunogenicity has been known as the main obstacle in the clinical use of RITs. Various strategies have been proposed to overcome these limitations, including immunosuppressive therapy, humanization of the antibody fragment moiety, generation of immunotoxins originated from endogenous human cytotoxic enzymes, and modification of the toxin moiety to escape the immune system. This paper is devoted to review recent advances in the design of immunotoxins with lower immunogenicity.
Collapse
Affiliation(s)
- Maryam Dashtiahangar
- Department of Biology, Faculty of Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Leila Rahbarnia
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safar Farajnia
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Arezoo Gowhari Shabgah
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Ghasemali
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
29
|
Hussein NH, Amin NS, El Tayebi HM. GPI-AP: Unraveling a New Class of Malignancy Mediators and Potential Immunotherapy Targets. Front Oncol 2020; 10:537311. [PMID: 33344222 PMCID: PMC7746843 DOI: 10.3389/fonc.2020.537311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/19/2020] [Indexed: 12/22/2022] Open
Abstract
With millions of cases diagnosed annually and high economic burden to cover expensive costs, cancer is one of the most difficult diseases to treat due to late diagnosis and severe adverse effects from conventional therapy. This creates an urgent need to find new targets for early diagnosis and therapy. Progress in research revealed the key steps of carcinogenesis. They are called cancer hallmarks. Zooming in, cancer hallmarks are characterized by ligands binding to their cognate receptor and so triggering signaling cascade within cell to make response for stimulus. Accordingly, understanding membrane topology is vital. In this review, we shall discuss one type of transmembrane proteins: Glycosylphosphatidylinositol-Anchored Proteins (GPI-APs), with specific emphasis on those involved in tumor cells by evading immune surveillance and future applications for diagnosis and immune targeted therapy.
Collapse
|
30
|
Shih TC, Wang L, Wang HC, Wan YJY. Glypican-3: A molecular marker for the detection and treatment of hepatocellular carcinoma ☆. LIVER RESEARCH 2020; 4:168-172. [PMID: 33384879 PMCID: PMC7771890 DOI: 10.1016/j.livres.2020.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with a fairly poor prognosis (5-year survival of less than 50%). Using sorafenib, the only food and drug administration (FDA)-approved drug, HCC cannot be effectively treated; it can only be controlled at most for a couple of months. There is a great need to develop efficacious treatment against this debilitating disease. Glypican-3 (GPC3), a member of the glypican family that attaches to the cell surface by a glycosylphosphatidylinositol anchor, is overexpressed in HCC cases and is elevated in the serum of a large proportion of patients with HCC. GPC3 expression contributes to HCC growth and metastasis. Furthermore, several different types of antibodies targeting GPC3 have been developed. The aim of this review is to summarize the current literatures on the GPC3 expression in human HCC, molecular mechanisms of GPC3 regulation and antibodies targeting GPC3.
Collapse
Affiliation(s)
- Tsung-Chieh Shih
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Lijun Wang
- Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA, USA
| | - Hsiao-Chi Wang
- Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Yu-Jui Yvonne Wan
- Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA, USA,Corresponding author. Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA, USA. (Y.-J.Y. Wan)
| |
Collapse
|
31
|
Fischer A, Wolf I, Fuchs H, Masilamani AP, Wolf P. Pseudomonas Exotoxin A Based Toxins Targeting Epidermal Growth Factor Receptor for the Treatment of Prostate Cancer. Toxins (Basel) 2020; 12:E753. [PMID: 33260619 PMCID: PMC7761469 DOI: 10.3390/toxins12120753] [Citation(s) in RCA: 5] [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: 09/30/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) was found to be a valuable target on prostate cancer (PCa) cells. However, EGFR inhibitors mostly failed in clinical studies with patients suffering from PCa. We therefore tested the targeted toxins EGF-PE40 and EGF-PE24mut consisting of the natural ligand EGF as binding domain and PE40, the natural toxin domain of Pseudomonas Exotoxin A, or PE24mut, the de-immunized variant thereof, as toxin domains. Both targeted toxins were expressed in the periplasm of E.coli and evoked an inhibition of protein biosynthesis in EGFR-expressing PCa cells. Concentration- and time-dependent killing of PCa cells was found with IC50 values after 48 and 72 h in the low nanomolar or picomolar range based on the induction of apoptosis. EGF-PE24mut was found to be about 11- to 120-fold less toxic than EGF-PE40. Both targeted toxins were more than 600 to 140,000-fold more cytotoxic than the EGFR inhibitor erlotinib. Due to their high and specific cytotoxicity, the EGF-based targeted toxins EGF-PE40 and EGF-PE24mut represent promising candidates for the future treatment of PCa.
Collapse
Affiliation(s)
- Alexandra Fischer
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (A.F.); (I.W.); (A.P.M.)
- Department of Urology, Antibody-Based Diagnostics and Therapies, Medical Center—University of Freiburg, Breisacher Str. 66, 79106 Freiburg, Germany
| | - Isis Wolf
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (A.F.); (I.W.); (A.P.M.)
- Department of Urology, Antibody-Based Diagnostics and Therapies, Medical Center—University of Freiburg, Breisacher Str. 66, 79106 Freiburg, Germany
| | - Hendrik Fuchs
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany;
| | - Anie Priscilla Masilamani
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (A.F.); (I.W.); (A.P.M.)
- Department of Urology, Antibody-Based Diagnostics and Therapies, Medical Center—University of Freiburg, Breisacher Str. 66, 79106 Freiburg, Germany
| | - Philipp Wolf
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (A.F.); (I.W.); (A.P.M.)
- Department of Urology, Antibody-Based Diagnostics and Therapies, Medical Center—University of Freiburg, Breisacher Str. 66, 79106 Freiburg, Germany
| |
Collapse
|
32
|
Shramova E, Proshkina G, Shipunova V, Ryabova A, Kamyshinsky R, Konevega A, Schulga A, Konovalova E, Telegin G, Deyev S. Dual Targeting of Cancer Cells with DARPin-Based Toxins for Overcoming Tumor Escape. Cancers (Basel) 2020; 12:cancers12103014. [PMID: 33081407 PMCID: PMC7602955 DOI: 10.3390/cancers12103014] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/06/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Targeted therapy of solid tumors represents a great challenge because of heterogeneity of tumor-associated antigen expression. To overcome this obstacle we propose a dual targeting therapy based on protein preparations capable of recognizing different of tumor-associated antigens on a tumor cell producing a directed cytotoxic effect. The dual specific therapy of breast carcinoma-bearing mice using the designed preparations eliminates both the primary tumor and distant metastases. The mono-targeting therapy aimed at single tumor-associated antigen did not suppress metastases at all. The proposed approach can serve as a potential therapeutic strategy that surpasses mono-specific targeting strategies in the anti-cancer efficacy. Abstract We report here a combined anti-cancer therapy directed toward HER2 and EpCAM, common tumor-associated antigens of breast cancer cells. The combined therapeutic effect is achieved owing to two highly toxic proteins—a low immunogenic variant of Pseudomonas aeruginosa exotoxin A and ribonuclease Barnase from Bacillus amyloliquefaciens. The delivery of toxins to cancer cells was carried out by targeting designed ankyrin repeat proteins (DARPins). We have shown that both target agents efficiently accumulate in the tumor. Simultaneous treatment of breast carcinoma-bearing mice with anti-EpCAM fusion toxin based on LoPE and HER2-specific liposomes loaded with Barnase leads to concurrent elimination of primary tumor and metastases. Monotherapy with anti-HER2- or anti-EpCAM-toxins did not produce a comparable effect on metastases. The proposed approach can be considered as a promising strategy for significant improvement of cancer therapy.
Collapse
Affiliation(s)
- Elena Shramova
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho–Maklaya Street 16/10, 117997 Moscow, Russia; (V.S.); (A.S.); (E.K.); (G.T.); (S.D.)
- Correspondence: (E.S.); (G.P.); Tel.: +7-9169503549 (E.S.); +7-9167997089 (G.P.)
| | - Galina Proshkina
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho–Maklaya Street 16/10, 117997 Moscow, Russia; (V.S.); (A.S.); (E.K.); (G.T.); (S.D.)
- Correspondence: (E.S.); (G.P.); Tel.: +7-9169503549 (E.S.); +7-9167997089 (G.P.)
| | - Victoria Shipunova
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho–Maklaya Street 16/10, 117997 Moscow, Russia; (V.S.); (A.S.); (E.K.); (G.T.); (S.D.)
| | - Anastasia Ryabova
- Prokhorov General Physics Institute, Russian Academy of Sciences, Vavilova Street 38, 119991 Moscow, Russia;
| | - Roman Kamyshinsky
- National Research Center “Kurchatov Institute”, Akademika Kurchatova pl. 1, 123182 Moscow, Russia; (R.K.); (A.K.)
- Shubnikov Institute of Crystallography of Federal Scientific Research Centre ‘Crystallography and Photonics’ of Russian Academy of Sciences, Leninskiy Prospect, 59, 119333 Moscow, Russia
- Moscow Institute of Physics and Technology, Institutsky Lane 9, Dolgoprudny, 141701 Moscow, Russia
| | - Andrey Konevega
- National Research Center “Kurchatov Institute”, Akademika Kurchatova pl. 1, 123182 Moscow, Russia; (R.K.); (A.K.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, Orlova Roscha 1, 188300 Gatchina, Russia
- Peter the Great St. Petersburg Polytechnic University, Politehnicheskaya 29, 195251 St. Petersburg, Russia
| | - Aleksey Schulga
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho–Maklaya Street 16/10, 117997 Moscow, Russia; (V.S.); (A.S.); (E.K.); (G.T.); (S.D.)
| | - Elena Konovalova
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho–Maklaya Street 16/10, 117997 Moscow, Russia; (V.S.); (A.S.); (E.K.); (G.T.); (S.D.)
| | - Georgij Telegin
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho–Maklaya Street 16/10, 117997 Moscow, Russia; (V.S.); (A.S.); (E.K.); (G.T.); (S.D.)
| | - Sergey Deyev
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho–Maklaya Street 16/10, 117997 Moscow, Russia; (V.S.); (A.S.); (E.K.); (G.T.); (S.D.)
- The Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| |
Collapse
|
33
|
Bera TK. Anti-BCMA Immunotoxins: Design, Production, and Preclinical Evaluation. Biomolecules 2020; 10:biom10101387. [PMID: 33003418 PMCID: PMC7600380 DOI: 10.3390/biom10101387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a B-cell malignancy that is incurable for a majority of patients. B-cell maturation antigen (BCMA) is a lineage-restricted differentiation protein highly expressed in multiple myeloma cells but not in other normal tissues except normal plasma B cells. Due to the restricted expression and being a cell surface membrane protein, BCMA is an ideal target for immunotherapy approaches in MM. Recombinant immunotoxins (RITs) are a novel class of protein therapeutics that are composed of the Fv or Fab portion of an antibody fused to a cytotoxic agent. RITs were produced by expressing plasmids encoding the components of the anti-BCMA RITs in E. coli followed by inclusion body preparation, solubilization, renaturation, and purification by column chromatography. The cytotoxic activity of RITs was tested in vitro by WST-8 assays using BCMA expressing cell lines and on cells isolated from MM patients. The in vivo efficacy of RITs was tested in a xenograft mouse model using BCMA expressing multiple myeloma cell lines. Anti-BCMA recombinant immunotoxins are very effective in killing myeloma cell lines and cells isolated from myeloma patients expressing BCMA. Two mouse models of myeloma showed that the anti-BCMA immunotoxins can produce a long-term complete response and warrant further preclinical development.
Collapse
Affiliation(s)
- Tapan K Bera
- Laboratory of Molecular Biology, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
34
|
UCNP-based Photoluminescent Nanomedicines for Targeted Imaging and Theranostics of Cancer. Molecules 2020; 25:molecules25184302. [PMID: 32961731 PMCID: PMC7571190 DOI: 10.3390/molecules25184302] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/13/2020] [Accepted: 09/17/2020] [Indexed: 12/22/2022] Open
Abstract
Theranostic approach is currently among the fastest growing trends in cancer treatment. It implies the creation of multifunctional agents for simultaneous precise diagnosis and targeted impact on tumor cells. A new type of theranostic complexes was created based on NaYF4: Yb,Tm upconversion nanoparticles coated with polyethylene glycol and functionalized with the HER2-specific recombinant targeted toxin DARPin-LoPE. The obtained agents bind to HER2-overexpressing human breast adenocarcinoma cells and demonstrate selective cytotoxicity against this type of cancer cells. Using fluorescent human breast adenocarcinoma xenograft models, the possibility of intravital visualization of the UCNP-based complexes biodistribution and accumulation in tumor was demonstrated.
Collapse
|
35
|
Liang S, Zhang C. Prediction of immunogenicity for humanized and full human therapeutic antibodies. PLoS One 2020; 15:e0238150. [PMID: 32866159 PMCID: PMC7458303 DOI: 10.1371/journal.pone.0238150] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/10/2020] [Indexed: 01/02/2023] Open
Abstract
Immunogenicity is an important concern for therapeutic antibodies during drug development. By analyzing co-crystal structures of idiotypic antibodies and their antibodies, we found that anti-idiotypic antibodies usually bind the Complementarity Determining Regions (CDR) of idiotypic antibodies. Sequence and structural features were identified for distinguishing immunogenic antibodies from non-immunogenic antibodies. For example, non-immunogenic antibodies have a significantly larger cavity volume at the CDR region and a more hydrophobic CDR-H3 loop than immunogenic antibodies. Antibodies containing no Gly at the turn of CDR-H2 loop are often immunogenic. We integrated these features together with a machine learning platform to Predict Immunogenicity for humanized and full human THerapeutic Antibodies (PITHA). This method achieved an accuracy of 83% in leave-one-out experiment for 29 therapeutic antibodies with available crystal structures. The accuracy decreased to 65% for 23 test antibodies with modeled structures, because their crystal structures were not available, and the prediction was made with modeled structures. The server of this method is accessible at http://mabmedicine.com/PITHA.
Collapse
Affiliation(s)
- Shide Liang
- Department of Research and Development, Bio-Thera Solutions, Guangzhou, P. R. China
- * E-mail: (SL); (CZ)
| | - Chi Zhang
- School of Biological Sciences, University of Nebraska, Lincoln, NE, United States of America
- * E-mail: (SL); (CZ)
| |
Collapse
|
36
|
32A9, a novel human antibody for designing an immunotoxin and CAR-T cells against glypican-3 in hepatocellular carcinoma. J Transl Med 2020; 18:295. [PMID: 32746924 PMCID: PMC7398316 DOI: 10.1186/s12967-020-02462-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 07/28/2020] [Indexed: 12/21/2022] Open
Abstract
Background Treatment of hepatocellular carcinoma (HCC) using antibody-based targeted therapies, such as antibody conjugates and chimeric antigen receptor T (CAR-T) cell therapy, shows potent antitumor efficacy. Glypican-3 (GPC3) is an emerging HCC therapeutic target; therefore, antibodies against GPC3 would be useful tools for developing immunotherapies for HCC. Methods We isolated a novel human monoclonal antibody, 32A9, by phage display technology. We determined specificity, affinity, epitope and anti-tumor activity of 32A9, and developed 32A9-based immunotherapy technologies for evaluating the potency of HCC treatment in vitro or in vivo. Results 32A9 recognized human GPC3 with potent affinity and specificity. The epitope of 32A9 was located in the region of the GPC3 protein core close to the modification sites of the HS chain and outside of the Wnt-binding site of GPC3. The 32A9 antibody significantly inhibited HCC xenograft tumor growth in vivo. We then pursued two 32A9-based immunotherapeutic strategies by constructing an immunotoxin and CAR-T cells. The 32A9 immunotoxin exhibited specific cytotoxicity to GPC3-positive cancer cells, while 32A9 CAR-T cells efficiently eliminated GPC3-positive HCC cells in vitro and caused HCC xenograft tumor regressions in vivo. Conclusions Our study provides a rationale for 32A9 as a promising GPC3-specific antibody candidate for HCC immunotherapy.
Collapse
|
37
|
Mesothelin-Targeted Recombinant Immunotoxins for Solid Tumors. Biomolecules 2020; 10:biom10070973. [PMID: 32605175 PMCID: PMC7408136 DOI: 10.3390/biom10070973] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
Mesothelin (MSLN) is a cell surface glycoprotein normally expressed only on serosal surfaces, and not found in the parenchyma of vital organs. Many solid tumors also express MSLN, including mesothelioma and pancreatic adenocarcinoma. Due to this favorable expression profile, MSLN represents a viable target for directed anti-neoplastic therapies, such as recombinant immunotoxins (iToxs). Pre-clinical testing of MSLN-targeted iTox’s has yielded a strong body of evidence for activity against a number of solid tumors. This has led to multiple clinical trials, testing the safety and efficacy of the clinical leads SS1P and LMB-100. While promising clinical results have been observed, neutralizing anti-drug antibody (ADA) formation presents a major challenge to overcome in the therapeutic development process. Additionally, on-target, off-tumor toxicity from serositis and non-specific capillary leak syndrome (CLS) also limits the dose, and therefore, impact anti-tumor activity. This review summarizes existing pre-clinical and clinical data on MSLN-targeted iTox’s. In addition, we address the potential future directions of research to enhance the activity of these anti-tumor agents.
Collapse
|
38
|
Mazor R, Pastan I. Immunogenicity of Immunotoxins Containing Pseudomonas Exotoxin A: Causes, Consequences, and Mitigation. Front Immunol 2020; 11:1261. [PMID: 32695104 PMCID: PMC7333791 DOI: 10.3389/fimmu.2020.01261] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/18/2020] [Indexed: 12/20/2022] Open
Abstract
Immunotoxins are cytolytic fusion proteins developed for cancer therapy, composed of an antibody fragment that binds to a cancer cell and a protein toxin fragment that kills the cell. Pseudomonas exotoxin A (PE) is a potent toxin that is used for the killing moiety in many immunotoxins. Moxetumomab Pasudotox (Lumoxiti) contains an anti-CD22 Fv and a 38 kDa portion of PE. Lumoxiti was discovered in the Laboratory of Molecular Biology at the U.S. National Cancer Institute and co-developed with Medimmune/AstraZeneca to treat hairy cell leukemia. In 2018 Lumoxiti was approved by the US Food and Drug Administration for the treatment of drug-resistant Hairy Cell Leukemia. Due to the bacterial origin of the killing moiety, immunotoxins containing PE are highly immunogenic in patients with normal immune systems, but less immunogenic in patients with hematologic malignancies, whose immune systems are often compromised. LMB-100 is a de-immunized variant of the toxin with a humanized antibody that targets mesothelin and a PE toxin that was rationally designed for diminished reactivity with antibodies and B cell receptors. It is now being evaluated in clinical trials for the treatment of mesothelioma and pancreatic cancer and is showing somewhat diminished immunogenicity compared to its un modified parental counterpart. Here we review the immunogenicity of the original and de-immunized PE immunotoxins in mice and patients, the development of anti-drug antibodies (ADAs), their impact on drug availability and their effect on clinical efficacy. Efforts to mitigate the immunogenicity of immunotoxins and its impact on immunogenicity will be described including rational design to identify, remove, or suppress B cell or T cell epitopes, and combination of immunotoxins with immune modulating drugs.
Collapse
Affiliation(s)
- Ronit Mazor
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
39
|
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: 1.4] [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
|
40
|
Bioinformatics Predictions, Expression, Purification and Structural Analysis of the PE38KDEL-scfv Immunotoxin Against EPHA2 Receptor. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-019-09901-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
41
|
Fleming BD, Urban DJ, Hall M, Longerich T, Greten T, Pastan I, Ho M. Engineered Anti-GPC3 Immunotoxin, HN3-ABD-T20, Produces Regression in Mouse Liver Cancer Xenografts Through Prolonged Serum Retention. Hepatology 2020; 71:1696-1711. [PMID: 31520528 PMCID: PMC7069773 DOI: 10.1002/hep.30949] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 09/08/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Treatment of hepatocellular carcinomas using our glypican-3 (GPC3)-targeting human nanobody (HN3) immunotoxins causes potent tumor regression by blocking protein synthesis and down-regulating the Wnt signaling pathway. However, immunogenicity and a short serum half-life may limit the ability of immunotoxins to transition to the clinic. APPROACH AND RESULTS To address these concerns, we engineered HN3-based immunotoxins to contain various deimmunized Pseudomonas exotoxin (PE) domains. This included HN3-T20, which was modified to remove T-cell epitopes and contains a PE domain II truncation. We compared them to our previously reported B-cell deimmunized immunotoxin (HN3-mPE24) and our original HN3-immunotoxin with a wild-type PE domain (HN3-PE38). All of our immunotoxins displayed high affinity to human GPC3, with HN3-T20 having a KD value of 7.4 nM. HN3-T20 retained 73% enzymatic activity when compared with the wild-type immunotoxin in an adenosine diphosphate-ribosylation assay. Interestingly, a real-time cell growth inhibition assay demonstrated that a single dose of HN3-T20 at 62.5 ng/mL (1.6 nM) was capable of inhibiting nearly all cell proliferation during the 10-day experiment. To enhance HN3-T20's serum retention, we tested the effect of adding a streptococcal albumin-binding domain (ABD) and a llama single-domain antibody fragment specific for mouse and human serum albumin. For the detection of immunotoxin in mouse serum, we developed a highly sensitive enzyme-linked immunosorbent assay and found that HN3-ABD-T20 had a 45-fold higher serum half-life than HN3-T20 (326 minutes vs. 7.3 minutes); consequently, addition of an ABD resulted in HN3-ABD-T20-mediated tumor regression at 1 mg/kg. CONCLUSION These data indicate that ABD-containing deimmunized HN3-T20 immunotoxins are high-potency therapeutics ready to be evaluated in clinical trials for the treatment of liver cancer.
Collapse
Affiliation(s)
- Bryan D. Fleming
- Laboratory of Molecular Biology, National Cancer Institute, Bethesda, Maryland, 20892
| | - Daniel J. Urban
- Chemical Genomics Center, National Center for Advancing Translational Sciences, Rockville, Maryland, 20850
| | - Matthew Hall
- Chemical Genomics Center, National Center for Advancing Translational Sciences, Rockville, Maryland, 20850
| | - Thomas Longerich
- Institute of Pathology, University Hospital, Heidelberg, Germany, 69120
| | - Tim Greten
- Thoracic and Gastrointestinal Oncology Branch, National Cancer Institute, Bethesda, Maryland, 20892
| | - Ira Pastan
- Laboratory of Molecular Biology, National Cancer Institute, Bethesda, Maryland, 20892
| | - Mitchell Ho
- Laboratory of Molecular Biology, National Cancer Institute, Bethesda, Maryland, 20892
| |
Collapse
|
42
|
HER2-Specific Pseudomonas Exotoxin A PE25 Based Fusions: Influence of Targeting Domain on Target Binding, Toxicity, and In Vivo Biodistribution. Pharmaceutics 2020; 12:pharmaceutics12040391. [PMID: 32344762 PMCID: PMC7238247 DOI: 10.3390/pharmaceutics12040391] [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: 03/27/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 11/23/2022] Open
Abstract
The human epidermal growth factor receptor 2 (HER2) is a clinically validated target for cancer therapy, and targeted therapies are often used in regimens for patients with a high HER2 expression level. Despite the success of current drugs, a number of patients succumb to their disease, which motivates development of novel drugs with other modes of action. We have previously shown that an albumin binding domain-derived affinity protein with specific affinity for HER2, ADAPT6, can be used to deliver the highly cytotoxic protein domain PE25, a derivative of Pseudomonas exotoxin A, to HER2 overexpressing malignant cells, leading to potent and specific cell killing. In this study we expanded the investigation for an optimal targeting domain and constructed two fusion toxins where a HER2-binding affibody molecule, ZHER2:2891, or the dual-HER2-binding hybrid ZHER2:2891-ADAPT6 were used for cancer cell targeting. We found that both targeting domains conferred strong binding to HER2; both to the purified extracellular domain and to the HER2 overexpressing cell line SKOV3. This resulted in fusion toxins with high cytotoxic potency toward cell lines with high expression levels of HER2, with EC50 values between 10 and 100 pM. For extension of the plasma half-life, an albumin binding domain was also included. Intravenous injection of the fusion toxins into mice showed a profound influence of the targeting domain on biodistribution. Compared to previous results, with ADAPT6 as targeting domain, ZHER2:2891 gave rise to further extension of the plasma half-life and also shifted the clearance route of the fusion toxin from the liver to the kidneys. Collectively, the results show that the targeting domain has a major impact on uptake of PE25-based fusion toxins in different organs. The results also show that PE25-based fusion toxins with high affinity to HER2 do not necessarily increase the cytotoxicity beyond a certain point in affinity. In conclusion, ZHER2:2891 has the most favorable characteristics as targeting domain for PE25.
Collapse
|
43
|
Lee S, Park S, Nguyen MT, Lee E, Kim J, Baek S, Kim CJ, Jang YJ, Choe H. A chemical conjugate between HER2-targeting antibody fragment and Pseudomonas exotoxin A fragment demonstrates cytotoxic effects on HER2-expressing breast cancer cells. BMB Rep 2020. [PMID: 30670149 PMCID: PMC6726212 DOI: 10.5483/bmbrep.2019.52.8.250] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Conventionally, immunotoxins have been produced as a single polypeptide from fused genes of an antibody fragment and a toxin. In this study, we adopted a unique approach of chemical conjugation of a toxin protein and an antibody fragment. The two genes were separately expressed in Escherichia coli and purified to high levels of purity. The two purified proteins were conjugated using a chemical linker. The advantage of this approach is its ability to overcome the problem of low recombinant immunotoxin production observed in some immunotoxins. Another advantage is that various combinations of immunotoxins can be prepared with fewer efforts, because the chemical conjugation of components is relatively simpler than the processes involved in cloning, expression, and purification of multiple immunotoxins. As a proof of concept, the scFv of trastuzumab and the PE24 fragment of Pseudomonas exotoxin A were separately produced using E. coli and then chemically crosslinked. The new immunotoxin was tested on four breast cancer cell lines variably expressing HER2. The chemically crosslinked immunotoxin exhibited cytotoxicity in proportion to the expression level of HER2. In conclusion, the present study revealed an alternative method of generating an immunotoxin that could effectively reduce the viability of HER2-expressing breast cancer cells. These results suggest the effectiveness of this method of immunotoxin crosslinking as a suitable alternative for producing immunotoxins. [BMB
Collapse
Affiliation(s)
- Sunju Lee
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Sangsu Park
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Minh Tan Nguyen
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea; Department of Biotechnology, NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
| | - Eunyoung Lee
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Julee Kim
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Sangki Baek
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Chong Jai Kim
- Department of Pathology, Asan-Minnesota Institute for Innovating Transplantation, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Yeon Jin Jang
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Han Choe
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| |
Collapse
|
44
|
Application of therapeutic protein-based fusion toxins. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0040-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
45
|
Cerise A, Bera TK, Liu X, Wei J, Pastan I. Anti-Mesothelin Recombinant Immunotoxin Therapy for Colorectal Cancer. Clin Colorectal Cancer 2019; 18:192-199.e1. [PMID: 31345777 PMCID: PMC8317202 DOI: 10.1016/j.clcc.2019.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/20/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Mesothelin (MSLN) is a cell surface glycoprotein expressed at a high level on many malignancies, including pancreatic adenocarcinoma, serous ovarian cancer, and epithelioid mesothelioma. MSLN-targeted recombinant immunotoxins (RITs) consist of an anti-MSLN Fv fused to the catalytic domain of Pseudomonas exotoxin A. Recent data has also shown that MSLN is expressed at clinically relevant levels on the surface of colorectal cancer (CRC). In this study, CRC cell lines were tested for MSLN expression and susceptibility to MSLN-targeted RITs. MATERIALS AND METHODS CRC cell lines were tested for membranous MSLN expression via flow cytometry. Cell lines expressing MSLN were tested by WST-8 cell viability assay for sensitivity to various RITs and chemotherapeutic agents. CRC cell line SW-48 was tested in a mouse model for response to RIT as a single agent or in combination with actinomycin D and oxaliplatin. RESULTS CRC cell lines were susceptible to anti-MSLN RITs at half maximal inhibitory concentration levels comparable with those previously described in pancreatic cancer cell lines. In a nude mouse model, MSLN-targeted RIT treatment of SW48 CRC tumors resulted in a significant decrease in tumor volume. Although combination therapy with standard of care chemotherapeutic oxaliplatin did not improve tumor regressions, combination therapy with actinomycin D resulted in > 90% tumor volume reduction with 50% complete regressions. CONCLUSIONS These data support the development of anti-MSLN RITs as well as other MSLN-targeted therapies for CRC.
Collapse
Affiliation(s)
- Adam Cerise
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Tapan K Bera
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Xiufen Liu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Junxia Wei
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
46
|
Choi Y, Furlon JM, Amos RB, Griswold KE, Bailey-Kellogg C. DisruPPI: structure-based computational redesign algorithm for protein binding disruption. Bioinformatics 2019; 34:i245-i253. [PMID: 29949961 PMCID: PMC6022686 DOI: 10.1093/bioinformatics/bty274] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Motivation Disruption of protein–protein interactions can mitigate antibody recognition of therapeutic proteins, yield monomeric forms of oligomeric proteins, and elucidate signaling mechanisms, among other applications. While designing affinity-enhancing mutations remains generally quite challenging, both statistically and physically based computational methods can precisely identify affinity-reducing mutations. In order to leverage this ability to design variants of a target protein with disrupted interactions, we developed the DisruPPI protein design method (DISRUpting Protein–Protein Interactions) to optimize combinations of mutations simultaneously for both disruption and stability, so that incorporated disruptive mutations do not inadvertently affect the target protein adversely. Results Two existing methods for predicting mutational effects on binding, FoldX and INT5, were demonstrated to be quite precise in selecting disruptive mutations from the SKEMPI and AB-Bind databases of experimentally determined changes in binding free energy. DisruPPI was implemented to use an INT5-based disruption score integrated with an AMBER-based stability assessment and was applied to disrupt protein interactions in a set of different targets representing diverse applications. In retrospective evaluation with three different case studies, comparison of DisruPPI-designed variants to published experimental data showed that DisruPPI was able to identify more diverse interaction-disrupting and stability-preserving variants more efficiently and effectively than previous approaches. In prospective application to an interaction between enhanced green fluorescent protein (EGFP) and a nanobody, DisruPPI was used to design five EGFP variants, all of which were shown to have significantly reduced nanobody binding while maintaining function and thermostability. This demonstrates that DisruPPI may be readily utilized for effective removal of known epitopes of therapeutically relevant proteins. Availability and implementation DisruPPI is implemented in the EpiSweep package, freely available under an academic use license. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Yoonjoo Choi
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jacob M Furlon
- Thayer School of Engineering, Dartmouth, Hanover, NH, USA
| | - Ryan B Amos
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Karl E Griswold
- Thayer School of Engineering, Dartmouth, Hanover, NH, USA.,Norris Cotton Cancer Center at Dartmouth, Lebanon, NH, USA.,Department of Biological Sciences, Dartmouth, Hanover, NH, USA
| | | |
Collapse
|
47
|
Lee S, Park S, Nguyen MT, Lee E, Kim J, Baek S, Kim CJ, Jang YJ, Choe H. A chemical conjugate between HER2-targeting antibody fragment and Pseudomonas exotoxin A fragment demonstrates cytotoxic effects on HER2-expressing breast cancer cells. BMB Rep 2019; 52:496-501. [PMID: 30670149 PMCID: PMC6726212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 11/11/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2023] Open
Abstract
Conventionally, immunotoxins have been produced as a single polypeptide from fused genes of an antibody fragment and a toxin. In this study, we adopted a unique approach of chemical conjugation of a toxin protein and an antibody fragment. The two genes were separately expressed in Escherichia coli and purified to high levels of purity. The two purified proteins were conjugated using a chemical linker. The advantage of this approach is its ability to overcome the problem of low recombinant immunotoxin production observed in some immunotoxins. Another advantage is that various combinations of immunotoxins can be prepared with fewer efforts, because the chemical conjugation of components is relatively simpler than the processes involved in cloning, expression, and purification of multiple immunotoxins. As a proof of concept, the scFv of trastuzumab and the PE24 fragment of Pseudomonas exotoxin A were separately produced using E. coli and then chemically crosslinked. The new immunotoxin was tested on four breast cancer cell lines variably expressing HER2. The chemically crosslinked immunotoxin exhibited cytotoxicity in proportion to the expression level of HER2. In conclusion, the present study revealed an alternative method of generating an immunotoxin that could effectively reduce the viability of HER2-expressing breast cancer cells. These results suggest the effectiveness of this method of immunotoxin crosslinking as a suitable alternative for producing immunotoxins. [BMB Reports 2019; 52(8): 496-501].
Collapse
Affiliation(s)
- Sunju Lee
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Sangsu Park
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Minh Tan Nguyen
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
- Department of Biotechnology, NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City 70000,
Vietnam
| | - Eunyoung Lee
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Julee Kim
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Sangki Baek
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Chong Jai Kim
- Department of Pathology, Asan-Minnesota Institute for Innovating Transplantation, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Yeon Jin Jang
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Han Choe
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| |
Collapse
|
48
|
Ibáñez-Pérez R, Guerrero-Ochoa P, Al-Wasaby S, Navarro R, Tapia-Galisteo A, De Miguel D, Gonzalo O, Conde B, Martínez-Lostao L, Hurtado-Guerrero R, Sanz L, Anel A. Anti-tumoral potential of a human granulysin-based, CEA-targeted cytolytic immunotoxin. Oncoimmunology 2019; 8:1641392. [PMID: 31646080 DOI: 10.1080/2162402x.2019.1641392] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/01/2019] [Accepted: 07/03/2019] [Indexed: 12/12/2022] Open
Abstract
Granulysin is a protein present in the granules of human cytotoxic T lymphocytes (CTL) and natural killer (NK) cells, with cytolytic activity against microbes and tumors. Previous work demonstrated the therapeutic effect of intratumoral injection of recombinant granulysin using in vivo models of breast cancer and multiple myeloma. In the present work we have developed a granulysin gene fusion to the anti-carcinoembryonic antigen (CEA/CEACAM5) single chain Fv antibody fragment MFE23. Both granulysin and the granulysin-based immunotoxin were expressed in Pichia pastoris. The immunotoxin specifically recognized CEA, purified or expressed on the cell surface. Moreover, the bioactivity of the immunotoxin against several CEA+ cell lines was higher than that of granulysin alone. Granulysin and the immunotoxin were tested as a treatment in in vivo xenograft models in athymic mice. When injected intratumorally, both granulysin and the immunotoxin were able to inhibit tumor growth. Furthermore, systemic administration of the immunotoxin demonstrated a decrease in tumor growth in a CEA+ tumor-bearing mouse model, whereas granulysin did not exhibit a therapeutic effect. This is the first granulysin-based immunotoxin and the present work constitutes the proof of concept of its therapeutic potential.
Collapse
Affiliation(s)
- Raquel Ibáñez-Pérez
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| | - Patricia Guerrero-Ochoa
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| | - Sameer Al-Wasaby
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| | - Rocío Navarro
- Molecular Immunology Unit, "Puerta de Hierro" University Hospital, Majadahonda, Madrid, Spain
| | - Antonio Tapia-Galisteo
- Molecular Immunology Unit, "Puerta de Hierro" University Hospital, Majadahonda, Madrid, Spain
| | - Diego De Miguel
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| | - Oscar Gonzalo
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| | - Blanca Conde
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| | - Luis Martínez-Lostao
- Immunology Department, "Lozano Blesa" University Clinical Hospital, Zaragoza, Spain
| | - Ramón Hurtado-Guerrero
- Biocomputation and Physics of Complex Systems Institute (BIFI), University of Zaragoza, Zaragoza, Spain.,ARAID Foundation, Zaragoza, Spain
| | - Laura Sanz
- Molecular Immunology Unit, "Puerta de Hierro" University Hospital, Majadahonda, Madrid, Spain
| | - Alberto Anel
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| |
Collapse
|
49
|
Liu H, Lindbo S, Ding H, Altai M, Garousi J, Orlova A, Tolmachev V, Hober S, Gräslund T. Potent and specific fusion toxins consisting of a HER2‑binding, ABD‑derived affinity protein, fused to truncated versions of Pseudomonas exotoxin A. Int J Oncol 2019; 55:309-319. [PMID: 31180549 DOI: 10.3892/ijo.2019.4814] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/22/2019] [Indexed: 11/05/2022] Open
Abstract
Fusion toxins consisting of an affinity protein fused to toxic polypeptides derived from Pseudomonas exotoxin A (ETA) are promising agents for targeted cancer therapy. In this study, we examined whether fusion toxins consisting of an albumin binding domain‑derived affinity protein (ADAPT) interacting with human epidermal growth factor receptor 2 (HER2), coupled to the ETA‑derived polypeptides PE38X8 or PE25, with or without an albumin binding domain (ABD) for half‑life extension, can be used for specific killing of HER2‑expressing cells. The fusion toxins could easily be expressed in a soluble form in Escherichia coli and purified to homogeneity. All constructs had strong affinity for HER2 (KD 10 to 26 nM) and no tendency for aggregation could be detected. The fusion toxins including the ABD showed strong interaction with human and mouse serum albumin [equilibrium dissociation constant (KD) 1 to 3 nM and 2 to 10 nM, respectively]. The in vitro investigation of the cytotoxic potential revealed IC50‑values in the picomolar range for cells expressing high levels of HER2. The specificity was also demonstrated, by showing that free HER2 receptors on the target cells are required for fusion toxin activity. In mice, the fusion toxins containing the ABD exhibited an appreciably longer time in circulation. The uptake was highest in liver and kidney. Fusion with PE25 was associated with the highest hepatic uptake. Collectively, the results suggest that fusion toxins consisting of ADAPTs and ETA‑derivatives are promising agents for targeted cancer therapy.
Collapse
Affiliation(s)
- Hao Liu
- Department of Protein Science, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden
| | - Sarah Lindbo
- Department of Protein Science, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden
| | - Haozhong Ding
- Department of Protein Science, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden
| | - Mohamed Altai
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Sophia Hober
- Department of Protein Science, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden
| |
Collapse
|
50
|
Lee BS, Lee Y, Park J, Jeong BS, Jo M, Jung ST, Yoo TH. Construction of an immunotoxin via site-specific conjugation of anti-Her2 IgG and engineered Pseudomonas exotoxin A. J Biol Eng 2019; 13:56. [PMID: 31285754 PMCID: PMC6588878 DOI: 10.1186/s13036-019-0188-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/12/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Immunotoxins consisting of a toxin from bacteria or plants and a targeting module have been developed as potent anti-cancer therapeutics. The majority of them, especially those in preclinical or clinical testing stages, are fusion proteins of a toxin and antibody fragment. Immunotoxins based on full-length antibodies are less studied, even though the fragment crystallizable (Fc) domain plays an important role in regulating the concentration of immunoglobulin G (IgG) in the serum and in antibody-mediated immune responses against pathogens. RESULTS We devised a method to site-specifically conjugate IgG and another protein using a cysteine residue introduced into the IgG and a bio-orthogonally reactive unnatural amino acid incorporated into the other protein. The human epidermal growth factor receptor 2 (Her2)-targeting IgG, trastuzumab, was engineered to have an unpaired cysteine in the heavy chain, and an unnatural amino acid with the azido group was incorporated into an engineered Pseudomonas exotoxin A (PE24). The two protein molecules were conjugated site-specifically using a bifunctional linker having dibenzocyclooctyne and maleimide groups. Binding to Her2 and interaction with various Fc receptors of trastuzumab were not affected by the conjugation with PE24. The trastuzumab-PE24 conjugate was cytotoxic to Her2-overexpressing cell lines, which involved the inhibition of cellular protein synthesis due to the modification of elongation factor-2. CONCLUSIONS We constructed the site-specifically conjugated immunotoxin based on IgG and PE24, which induced target-specific cytotoxicity. To evaluate the molecule as a cancer therapeutic, animal studies are planned to assess tumor regression, half-life in blood, and in vivo immunogenicity. In addition, we expect that the site-specific conjugation method can be used to develop other antibody-protein conjugates for applications in therapeutics and diagnostics.
Collapse
Affiliation(s)
- Byeong Sung Lee
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 16499 South Korea
| | - Yumi Lee
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 16499 South Korea
| | - Jisoo Park
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 16499 South Korea
| | - Bo Seok Jeong
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 16499 South Korea
| | - Migyeong Jo
- Department of Applied Chemistry, Kookmin University, 77 Jeongneung-ro, Seongbuk-gu, Seoul, 02707 South Korea
| | - Sang Taek Jung
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, Seongbuk-gu, Seoul, 02841 South Korea
| | - Tae Hyeon Yoo
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 16499 South Korea
- Department of Applied Chemistry and Biological Engineering, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 16499 South Korea
| |
Collapse
|