1
|
Floudas CS, Sarkizova S, Ceccarelli M, Zheng W. Leveraging mRNA technology for antigen based immuno-oncology therapies. J Immunother Cancer 2025; 13:e010569. [PMID: 39848687 PMCID: PMC11784169 DOI: 10.1136/jitc-2024-010569] [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: 09/16/2024] [Accepted: 01/03/2025] [Indexed: 01/25/2025] Open
Abstract
The application of messenger RNA (mRNA) technology in antigen-based immuno-oncology therapies represents a significant advancement in cancer treatment. Cancer vaccines are an effective combinatorial partner to sensitize the host immune system to the tumor and boost the efficacy of immune therapies. Selecting suitable tumor antigens is the key step to devising effective vaccinations and amplifying the immune response. Tumor neoantigens are de novo epitopes derived from somatic mutations, avoiding T-cell central tolerance of self-epitopes and inducing immune responses to tumors. The identification and prioritization of patient-specific tumor neoantigens are based on advanced computational algorithms taking advantage of the profiling with next-generation sequencing considering factors involved in human leukocyte antigen (HLA)-peptide-T-cell receptor (TCR) complex formation, including peptide presentation, HLA-peptide affinity, and TCR recognition. This review discusses the development and clinical application of mRNA vaccines in oncology, with a particular focus on recent clinical trials and the computational workflows and methodologies for identifying both shared and individual antigens. While this review centers on therapeutic mRNA vaccines targeting existing tumors, it does not cover preventative vaccines. Preclinical experimental validations are crucial in cancer vaccine development, but we emphasize the computational approaches that facilitate neoantigen selection and design, highlighting their role in advancing mRNA vaccine development. The versatility and rapid development potential of mRNA make it an ideal platform for personalized neoantigen immunotherapy. We explore various strategies for antigen target identification, including tumor-associated and tumor-specific antigens and the computational tools used to predict epitopes capable of eliciting strong immune responses. We address key design considerations for enhancing the immunogenicity and stability of mRNA vaccines, as well as emerging trends and challenges in the field. This comprehensive overview highlights the therapeutic potential of mRNA-based cancer vaccines and underscores ongoing research efforts aimed at optimizing these therapies for improved clinical outcomes.
Collapse
Affiliation(s)
- Charalampos S Floudas
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | | | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center, Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Wei Zheng
- Moderna, Inc, Cambridge, Massachusetts, USA
| |
Collapse
|
2
|
Raghavan R, Friedrich MJ, King I, Chau-Duy-Tam Vo S, Strebinger D, Lash B, Kilian M, Platten M, Macrae RK, Song Y, Nivon L, Zhang F. Rational engineering of minimally immunogenic nucleases for gene therapy. Nat Commun 2025; 16:105. [PMID: 39747875 PMCID: PMC11696374 DOI: 10.1038/s41467-024-55522-1] [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: 08/07/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025] Open
Abstract
Genome editing using CRISPR-Cas systems is a promising avenue for the treatment of genetic diseases. However, cellular and humoral immunogenicity of genome editing tools, which originate from bacteria, complicates their clinical use. Here we report reduced immunogenicity (Red)(i)-variants of two clinically relevant nucleases, SaCas9 and AsCas12a. Through MHC-associated peptide proteomics (MAPPs) analysis, we identify putative immunogenic epitopes on each nuclease. Using computational modeling, we rationally design these proteins to evade the immune response. SaCas9 and AsCas12a Redi variants are substantially less recognized by adaptive immune components, including reduced binding affinity to MHC molecules and attenuated generation of cytotoxic T cell responses, yet maintain wild-type levels of activity and specificity. In vivo editing of PCSK9 with SaCas9.Redi.1 is comparable in efficiency to wild-type SaCas9, but significantly reduces undesired immune responses. This demonstrates the utility of this approach in engineering proteins to evade immune detection.
Collapse
Affiliation(s)
- Rumya Raghavan
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- McGovern Institute for Brain Research at MIT, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Howard Hughes Medical Institute, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Massachusetts, 02139, Cambridge, USA
| | - Mirco J Friedrich
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- McGovern Institute for Brain Research at MIT, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Howard Hughes Medical Institute, Cambridge, MA, 02139, USA
| | - Indigo King
- Cyrus Biotechnology, Seattle, WA, 98121, USA
| | - Samuel Chau-Duy-Tam Vo
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- McGovern Institute for Brain Research at MIT, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Howard Hughes Medical Institute, Cambridge, MA, 02139, USA
| | - Daniel Strebinger
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- McGovern Institute for Brain Research at MIT, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Howard Hughes Medical Institute, Cambridge, MA, 02139, USA
| | - Blake Lash
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- McGovern Institute for Brain Research at MIT, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Howard Hughes Medical Institute, Cambridge, MA, 02139, USA
| | - Michael Kilian
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Michael Platten
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Rhiannon K Macrae
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- McGovern Institute for Brain Research at MIT, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Howard Hughes Medical Institute, Cambridge, MA, 02139, USA
| | - Yifan Song
- Cyrus Biotechnology, Seattle, WA, 98121, USA
| | - Lucas Nivon
- Cyrus Biotechnology, Seattle, WA, 98121, USA
| | - Feng Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
- McGovern Institute for Brain Research at MIT, Cambridge, MA, 02139, USA.
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Howard Hughes Medical Institute, Cambridge, MA, 02139, USA.
| |
Collapse
|
3
|
Tong M, Palmer N, Dailamy A, Kumar A, Khaliq H, Han S, Finburgh E, Wing M, Hong C, Xiang Y, Miyasaki K, Portell A, Rainaldi J, Suhardjo A, Nourreddine S, Chew WL, Kwon EJ, Mali P. Robust genome and cell engineering via in vitro and in situ circularized RNAs. Nat Biomed Eng 2025; 9:109-126. [PMID: 39187662 DOI: 10.1038/s41551-024-01245-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 07/24/2024] [Indexed: 08/28/2024]
Abstract
Circularization can improve RNA persistence, yet simple and scalable approaches to achieve this are lacking. Here we report two methods that facilitate the pursuit of circular RNAs (cRNAs): cRNAs developed via in vitro circularization using group II introns, and cRNAs developed via in-cell circularization by the ubiquitously expressed RtcB protein. We also report simple purification protocols that enable high cRNA yields (40-75%) while maintaining low immune responses. These methods and protocols facilitate a broad range of applications in stem cell engineering as well as robust genome and epigenome targeting via zinc finger proteins and CRISPR-Cas9. Notably, cRNAs bearing the encephalomyocarditis internal ribosome entry enabled robust expression and persistence compared with linear capped RNAs in cardiomyocytes and neurons, which highlights the utility of cRNAs in these non-dividing cells. We also describe genome targeting via deimmunized Cas9 delivered as cRNA and a long-range multiplexed protein engineering methodology for the combinatorial screening of deimmunized protein variants that enables compatibility between persistence of expression and immunogenicity in cRNA-delivered proteins. The cRNA toolset will aid research and the development of therapeutics.
Collapse
Affiliation(s)
- Michael Tong
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Nathan Palmer
- Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Amir Dailamy
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Aditya Kumar
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Hammza Khaliq
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Sangwoo Han
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Emma Finburgh
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Madeleine Wing
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Camilla Hong
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Yichen Xiang
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Katelyn Miyasaki
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Andrew Portell
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Joseph Rainaldi
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Amanda Suhardjo
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Sami Nourreddine
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Wei Leong Chew
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Ester J Kwon
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Prashant Mali
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
4
|
Xu Z, Song J, Zhang H, Wei Z, Wei D, Yang G, Demongeot J, Zeng Q. A mathematical model simulating the adaptive immune response in various vaccines and vaccination strategies. Sci Rep 2024; 14:23995. [PMID: 39402093 PMCID: PMC11473516 DOI: 10.1038/s41598-024-74221-x] [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: 07/21/2024] [Accepted: 09/24/2024] [Indexed: 10/17/2024] Open
Abstract
Vaccination has been widely recognized as an effective measure for preventing infectious diseases. To facilitate quantitative research into the activation of adaptive immune responses in the human body by vaccines, it is important to develop an appropriate mathematical model, which can provide valuable guidance for vaccine development. In this study, we constructed a novel mathematical model to simulate the dynamics of antibody levels following vaccination, based on principles from immunology. Our model offers a concise and accurate representation of the kinetics of antibody response. We conducted a comparative analysis of antibody dynamics within the body after administering several common vaccines, including traditional inactivated vaccines, mRNA vaccines, and future attenuated vaccines based on defective interfering viral particles (DVG). Our findings suggest that booster shots play a crucial role in enhancing Immunoglobulin G (IgG) antibody levels, and we provide a detailed discussion on the advantages and disadvantages of different vaccine types. From a mathematical standpoint, our model proposes four essential approaches to guide vaccine design: enhancing antigenic T-cell immunogenicity, directing the production of high-affinity antibodies, reducing the rate of IgG decay, and lowering the peak level of vaccine antigen-antibody complexes. Our study contributes to the understanding of vaccine design and its application by explaining various phenomena and providing guidance in comprehending the interactions between antibodies and antigens during the immune process.
Collapse
Affiliation(s)
- Zhaobin Xu
- Department of Life Science, Dezhou University, Dezhou, 253023, China.
| | - Jian Song
- Department of Life Science, Dezhou University, Dezhou, 253023, China
| | - Hongmei Zhang
- Department of Life Science, Dezhou University, Dezhou, 253023, China
| | - Zhenlin Wei
- Department of Life Science, Dezhou University, Dezhou, 253023, China
| | - Dongqing Wei
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Meixi, Nanyang, 473006, Henan, P. R. China
- Peng Cheng National Laboratory, Vanke Cloud City Phase I Building 8, Xili Street, Nashan District, 518055, Shenzhen, Guangdong, P. R. China
| | - Guangyu Yang
- Department of Arts, Dezhou University, 253023, Dezhou, China
| | - Jacques Demongeot
- Laboratory AGEIS EA 7407, Team Tools for e-Gnosis Medical, Faculty of Medicine, University Grenoble Alpes (UGA), 38700, La Tronche, France.
| | - Qiangcheng Zeng
- Department of Life Science, Dezhou University, Dezhou, 253023, China.
| |
Collapse
|
5
|
Skeeters S, Bagale K, Stepanyuk G, Thieker D, Aguhob A, Chan KK, Dutzar B, Shalygin S, Shajahan A, Yang X, DaRosa PA, Frazier E, Sauer MM, Bogatzki L, Byrnes-Blake KA, Song Y, Azadi P, Tarcha E, Zhang L, Procko E. Modulation of the pharmacokinetics of soluble ACE2 decoy receptors through glycosylation. Mol Ther Methods Clin Dev 2024; 32:101301. [PMID: 39185275 PMCID: PMC11342882 DOI: 10.1016/j.omtm.2024.101301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 07/16/2024] [Indexed: 08/27/2024]
Abstract
The Spike of SARS-CoV-2 recognizes a transmembrane protease, angiotensin-converting enzyme 2 (ACE2), on host cells to initiate infection. Soluble derivatives of ACE2, in which Spike affinity is enhanced and the protein is fused to Fc of an immunoglobulin, are potent decoy receptors that reduce disease in animal models of COVID-19. Mutations were introduced into an ACE2 decoy receptor, including adding custom N-glycosylation sites and a cavity-filling substitution together with Fc modifications, which increased the decoy's catalytic activity and provided small to moderate enhancements of pharmacokinetics following intravenous and subcutaneous administration in humanized FcRn mice. Most prominently, sialylation of native glycans increases exposures by orders of magnitude, and the optimized decoy is therapeutically efficacious in a mouse COVID-19 model. Ultimately, an engineered and highly sialylated decoy receptor produced using methods suitable for manufacture of representative drug substance has high exposure with a 5- to 9-day half-life. Finally, peptide epitopes at mutated sites in the decoys generally have low binding to common HLA class II alleles and the predicted immunogenicity risk is low. Overall, glycosylation is a critical molecular attribute of ACE2 decoy receptors and modifications that combine tighter blocking of Spike with enhanced pharmacokinetics elevate this class of molecules as viable drug candidates.
Collapse
Affiliation(s)
| | - Kamal Bagale
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | - Sergei Shalygin
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Asif Shajahan
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Xu Yang
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | | | | | | | | | | | - Yifan Song
- Cyrus Biotechnology, Seattle, WA 98121, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | | | - Lianghui Zhang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Erik Procko
- Cyrus Biotechnology, Seattle, WA 98121, USA
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
6
|
Tusé D, McNulty M, McDonald KA, Buchman LW. A review and outlook on expression of animal proteins in plants. FRONTIERS IN PLANT SCIENCE 2024; 15:1426239. [PMID: 39239203 PMCID: PMC11374769 DOI: 10.3389/fpls.2024.1426239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/25/2024] [Indexed: 09/07/2024]
Abstract
This review delves into the multifaceted technologies, benefits and considerations surrounding the expression of animal proteins in plants, emphasizing its potential role in advancing global nutrition, enhancing sustainability, while being mindful of the safety considerations. As the world's population continues to grow and is projected to reach 9 billion people by 2050, there is a growing need for alternative protein sources that can meet nutritional demands while minimizing environmental impact. Plant expression of animal proteins is a cutting-edge biotechnology approach that allows crops to produce proteins traditionally derived from animals, offering a sustainable and resource-efficient manner of producing these proteins that diversifies protein production and increases food security. In the United States, it will be important for there to be clear guidance in order for these technologies to reach consumers. As consumer demand for sustainable and alternative food sources rise, biotechnologies can offer economic opportunities, making this emerging technology a key player in the market landscape.
Collapse
Affiliation(s)
- Daniel Tusé
- DT/Consulting Group, Sacramento, CA, United States
| | - Matthew McNulty
- Center for Cellular Agriculture, Tufts University, Medford, MA, United States
| | - Karen A McDonald
- Department of Chemical Engineering and Global Healthshare Initiative, University of California, Davis, Davis, CA, United States
| | - Leah W Buchman
- Biotechniology Innovation Organization, Agriculture and Environment, Washington, DC, United States
| |
Collapse
|
7
|
Meador K, Castells-Graells R, Aguirre R, Sawaya MR, Arbing MA, Sherman T, Senarathne C, Yeates TO. A suite of designed protein cages using machine learning and protein fragment-based protocols. Structure 2024; 32:751-765.e11. [PMID: 38513658 PMCID: PMC11162342 DOI: 10.1016/j.str.2024.02.017] [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: 10/19/2023] [Revised: 01/22/2024] [Accepted: 02/23/2024] [Indexed: 03/23/2024]
Abstract
Designed protein cages and related materials provide unique opportunities for applications in biotechnology and medicine, but their creation remains challenging. Here, we apply computational approaches to design a suite of tetrahedrally symmetric, self-assembling protein cages. For the generation of docked conformations, we emphasize a protein fragment-based approach, while for sequence design of the de novo interface, a comparison of knowledge-based and machine learning protocols highlights the power and increased experimental success achieved using ProteinMPNN. An analysis of design outcomes provides insights for improving interface design protocols, including prioritizing fragment-based motifs, balancing interface hydrophobicity and polarity, and identifying preferred polar contact patterns. In all, we report five structures for seven protein cages, along with two structures of intermediate assemblies, with the highest resolution reaching 2.0 Å using cryo-EM. This set of designed cages adds substantially to the body of available protein nanoparticles, and to methodologies for their creation.
Collapse
Affiliation(s)
- Kyle Meador
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | | | - Roman Aguirre
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Michael R Sawaya
- UCLA-DOE Institute for Genomics and Proteomics, Los Angeles, CA 90095, USA
| | - Mark A Arbing
- UCLA-DOE Institute for Genomics and Proteomics, Los Angeles, CA 90095, USA
| | - Trent Sherman
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Chethaka Senarathne
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Todd O Yeates
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA; UCLA-DOE Institute for Genomics and Proteomics, Los Angeles, CA 90095, USA.
| |
Collapse
|
8
|
Aoki K, Higashi K, Oda S, Manabe A, Maeda K, Morise J, Oka S, Inuki S, Ohno H, Oishi S, Nonaka M. Engineering a Low-Immunogenic Mirror-Image VHH against Vascular Endothelial Growth Factor. ACS Chem Biol 2024; 19:1194-1205. [PMID: 38695546 DOI: 10.1021/acschembio.4c00197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Immunogenicity is a major caveat of protein therapeutics. In particular, the long-term administration of protein therapeutic agents leads to the generation of antidrug antibodies (ADAs), which reduce drug efficacy while eliciting adverse events. One promising solution to this issue is the use of mirror-image proteins consisting of d-amino acids, which are resistant to proteolytic degradation in immune cells. We have recently reported the chemical synthesis of the enantiomeric form of the variable domain of the antibody heavy chain (d-VHH). However, identifying mirror-image antibodies capable of binding to natural ligands remains challenging. In this study, we developed a novel screening platform to identify a d-VHH specific for vascular endothelial growth factor A (VEGF-A). We performed mirror-image screening of two newly constructed synthetic VHH libraries displayed on T7 phage and identified VHH sequences that effectively bound to the mirror-image VEGF-A target (d-VEGF-A). We subsequently synthesized a d-VHH candidate that preferentially bound the native VEGF-A (l-VEGF-A) with submicromolar affinity. Furthermore, immunization studies in mice demonstrated that this d-VHH elicited no ADAs, unlike its corresponding l-VHH. Our findings highlight the utility of this novel d-VHH screening platform in the development of protein therapeutics exhibiting both reduced immunogenicity and improved efficacy.
Collapse
Affiliation(s)
- Keisuke Aoki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Laboratory of Medicinal Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8412, Japan
| | - Katsuaki Higashi
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Sakiho Oda
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Asako Manabe
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kayuu Maeda
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Jyoji Morise
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shogo Oka
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shinsuke Inuki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroaki Ohno
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shinya Oishi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Laboratory of Medicinal Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8412, Japan
| | - Motohiro Nonaka
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
9
|
Bhatt B, García-Díaz P, Foight GW. Synthetic transcription factor engineering for cell and gene therapy. Trends Biotechnol 2024; 42:449-463. [PMID: 37865540 DOI: 10.1016/j.tibtech.2023.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/23/2023]
Abstract
Synthetic transcription factors (synTFs) that control beneficial transgene expression are an important method to increase the safety and efficacy of cell and gene therapy. Reliance on synTF components from non-human sources has slowed progress in the field because of concerns about immunogenicity and inducer drug properties. Recent advances in human-derived DNA-binding domains (DBDs) and transcriptional activation domains (TADs) paired with novel control modules responsive to clinically approved small molecules have poised the synTF field to overcome these hurdles. Advances include controllers inducible by autonomous signaling inputs and more complex, multi-input synTF circuits. Demonstrations of advanced control strategies with human-derived transcription factor components in clinically relevant vectors and in vivo models will facilitate progression into the clinic.
Collapse
Affiliation(s)
- Bhoomi Bhatt
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, and Baylor College of Medicine, Houston, TX, USA
| | - Pablo García-Díaz
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, and Baylor College of Medicine, Houston, TX, USA
| | - Glenna Wink Foight
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, and Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
10
|
Thrift WJ, Perera J, Cohen S, Lounsbury NW, Gurung HR, Rose CM, Chen J, Jhunjhunwala S, Liu K. Graph-pMHC: graph neural network approach to MHC class II peptide presentation and antibody immunogenicity. Brief Bioinform 2024; 25:bbae123. [PMID: 38555476 PMCID: PMC10981672 DOI: 10.1093/bib/bbae123] [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: 01/02/2024] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 04/02/2024] Open
Abstract
Antigen presentation on MHC class II (pMHCII presentation) plays an essential role in the adaptive immune response to extracellular pathogens and cancerous cells. But it can also reduce the efficacy of large-molecule drugs by triggering an anti-drug response. Significant progress has been made in pMHCII presentation modeling due to the collection of large-scale pMHC mass spectrometry datasets (ligandomes) and advances in machine learning. Here, we develop graph-pMHC, a graph neural network approach to predict pMHCII presentation. We derive adjacency matrices for pMHCII using Alphafold2-multimer and address the peptide-MHC binding groove alignment problem with a simple graph enumeration strategy. We demonstrate that graph-pMHC dramatically outperforms methods with suboptimal inductive biases, such as the multilayer-perceptron-based NetMHCIIpan-4.0 (+20.17% absolute average precision). Finally, we create an antibody drug immunogenicity dataset from clinical trial data and develop a method for measuring anti-antibody immunogenicity risk using pMHCII presentation models. Our model increases receiver operating characteristic curve (ROC)-area under the ROC curve (AUC) by 2.57% compared to just filtering peptides by hits in OASis alone for predicting antibody drug immunogenicity.
Collapse
Affiliation(s)
| | - Jason Perera
- Genentech, 1 DNA Way, South San Francisco, California 94080, USA
| | - Sivan Cohen
- Genentech, 1 DNA Way, South San Francisco, California 94080, USA
| | | | - Hem R Gurung
- Genentech, 1 DNA Way, South San Francisco, California 94080, USA
| | | | - Jieming Chen
- Genentech, 1 DNA Way, South San Francisco, California 94080, USA
| | | | - Kai Liu
- Genentech, 1 DNA Way, South San Francisco, California 94080, USA
| |
Collapse
|
11
|
Aoki K, Manabe A, Kimura H, Katoh Y, Inuki S, Ohno H, Nonaka M, Oishi S. Mirror-Image Single-Domain Antibody for a Novel Nonimmunogenic Drug Scaffold. Bioconjug Chem 2023; 34:2055-2065. [PMID: 37883660 DOI: 10.1021/acs.bioconjchem.3c00372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Immunogenic responses by protein therapeutics often lead to reduced therapeutic effects and/or adverse effects via the generation of neutralizing antibodies and/or antidrug antibodies (ADA). Mirror-image proteins of the variable domain of the heavy chain of the heavy chain antibody (VHH) are potential novel protein therapeutics with high-affinity binding to target proteins and reduced immunogenicity because these mirror-image VHHs (d-VHHs) are less susceptible to proteolytic degradation in antigen-presenting cells (APCs). In this study, we investigated the preparation protocols of d-VHHs and their biological properties, including stereoselective target binding and immunogenicity. Initially, we established a facile synthetic process of two model VHHs [anti-GFP VHH and PMP12A2h1 (monomeric VHH of caplacizumab)] and their mirror-image proteins by three-step native chemical ligations (NCLs) from four peptide segments. The folded synthetic VHHs (l-anti-GFP VHH and l-PMP12A2h1) bound to the target proteins (EGFP and vWF-A1 domain, respectively), while their mirror-image proteins (d-anti-GFP VHH and d-PMP12A2h1) showed no binding to the native proteins. For biodistribution studies, l-VHH and d-VHH with single radioactive indium diethylenetriamine-pentaacid (111In-DTPA) labeling at the C-terminus were designed and synthesized by the established protocol. The distribution profiles were essentially similar between l-VHH and d-VHH, in which the probes accumulated in the kidney within 15 min after intravenous administration in mice, because of the small molecular size of VHHs. Comparative assessment of the immunogenicity responses revealed that d-VHH-induced levels of ADA generation were significantly lower than those of native VHH, regardless of the peptide sequences and administration routes. The resulting scaffold investigated should be applicable in the design of d-VHHs with various C-terminal CDR3 sequences, which can be identified by screening using display technologies.
Collapse
Affiliation(s)
- Keisuke Aoki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Laboratory of Medicinal Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8412, Japan
| | - Asako Manabe
- Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hiroyuki Kimura
- Laboratory of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Yohei Katoh
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shinsuke Inuki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroaki Ohno
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Motohiro Nonaka
- Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shinya Oishi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Laboratory of Medicinal Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8412, Japan
| |
Collapse
|
12
|
Shishparenok AN, Gladilina YA, Zhdanov DD. Engineering and Expression Strategies for Optimization of L-Asparaginase Development and Production. Int J Mol Sci 2023; 24:15220. [PMID: 37894901 PMCID: PMC10607044 DOI: 10.3390/ijms242015220] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Genetic engineering for heterologous expression has advanced in recent years. Model systems such as Escherichia coli, Bacillus subtilis and Pichia pastoris are often used as host microorganisms for the enzymatic production of L-asparaginase, an enzyme widely used in the clinic for the treatment of leukemia and in bakeries for the reduction of acrylamide. Newly developed recombinant L-asparaginase (L-ASNase) may have a low affinity for asparagine, reduced catalytic activity, low stability, and increased glutaminase activity or immunogenicity. Some successful commercial preparations of L-ASNase are now available. Therefore, obtaining novel L-ASNases with improved properties suitable for food or clinical applications remains a challenge. The combination of rational design and/or directed evolution and heterologous expression has been used to create enzymes with desired characteristics. Computer design, combined with other methods, could make it possible to generate mutant libraries of novel L-ASNases without costly and time-consuming efforts. In this review, we summarize the strategies and approaches for obtaining and developing L-ASNase with improved properties.
Collapse
Affiliation(s)
- Anastasiya N. Shishparenok
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (A.N.S.); (Y.A.G.)
| | - Yulia A. Gladilina
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (A.N.S.); (Y.A.G.)
| | - Dmitry D. Zhdanov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (A.N.S.); (Y.A.G.)
- Department of Biochemistry, Peoples’ Friendship University of Russia named after Patrice Lumumba (RUDN University), Miklukho—Maklaya St. 6, 117198 Moscow, Russia
| |
Collapse
|
13
|
Meador K, Castells-Graells R, Aguirre R, Sawaya MR, Arbing MA, Sherman T, Senarathne C, Yeates TO. A Suite of Designed Protein Cages Using Machine Learning Algorithms and Protein Fragment-Based Protocols. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.09.561468. [PMID: 37873110 PMCID: PMC10592684 DOI: 10.1101/2023.10.09.561468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Designed protein cages and related materials provide unique opportunities for applications in biotechnology and medicine, while methods for their creation remain challenging and unpredictable. In the present study, we apply new computational approaches to design a suite of new tetrahedrally symmetric, self-assembling protein cages. For the generation of docked poses, we emphasize a protein fragment-based approach, while for de novo interface design, a comparison of computational protocols highlights the power and increased experimental success achieved using the machine learning program ProteinMPNN. In relating information from docking and design, we observe that agreement between fragment-based sequence preferences and ProteinMPNN sequence inference correlates with experimental success. Additional insights for designing polar interactions are highlighted by experimentally testing larger and more polar interfaces. In all, using X-ray crystallography and cryo-EM, we report five structures for seven protein cages, with atomic resolution in the best case reaching 2.0 Å. We also report structures of two incompletely assembled protein cages, providing unique insights into one type of assembly failure. The new set of designed cages and their structures add substantially to the body of available protein nanoparticles, and to methodologies for their creation.
Collapse
Affiliation(s)
- Kyle Meador
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA 90095
| | | | - Roman Aguirre
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA 90095
| | - Michael R. Sawaya
- UCLA-DOE Institute for Genomics and Proteomics, Los Angeles, CA, USA 90095
| | - Mark A. Arbing
- UCLA-DOE Institute for Genomics and Proteomics, Los Angeles, CA, USA 90095
| | - Trent Sherman
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA 90095
| | - Chethaka Senarathne
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA 90095
| | - Todd O. Yeates
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA 90095
- UCLA-DOE Institute for Genomics and Proteomics, Los Angeles, CA, USA 90095
| |
Collapse
|
14
|
Palacio-Castañeda V, van de Crommert B, Verploegen E, Overeem M, van Oostrum J, Verdurmen WP. Potent and selective eradication of tumor cells by an EpCAM-targeted Ras-degrading enzyme. Mol Ther Oncolytics 2023; 30:16-26. [PMID: 37485031 PMCID: PMC10362089 DOI: 10.1016/j.omto.2023.06.002] [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: 12/22/2022] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
Despite decades of efforts, an urgent need remains to develop tumor cell-selective rat sarcoma (Ras)-targeting therapies that can treat patients with Ras-driven tumors. Here we report modular engineered proteins that degrade Ras selectively in tumor cells that overexpress the tumor cell marker epithelial cell adhesion molecule (EpCAM) by fusing the Ras degrader Ras-Rap1-specific endopeptidase with the translocation domain of the Pseudomonas aeruginosa exotoxin A (ETA) or diphtheria toxin (DT). Redirection to EpCAM is achieved by a designed ankyrin repeat protein. In two-dimensional tumor cell cultures, complete degradation of Ras proteins after 24 h was observed with EpCAM-targeted Ras degraders fused to ETA or DT in EpCAM-overexpressing MCF7 and HCT116 cells, with median inhibition concentration values at sub-nanomolar levels. The viability of EpCAM-low non-cancerous fibroblasts remained unaffected. In a three-dimensional (3D) tumor-on-a-chip system that mimics the natural tumor microenvironment, effective Ras degradation and selective toxicity toward tumor cells, particularly with the ETA-fused constructs, was determined on-chip. To conclude, we demonstrate the potential of modular engineered proteins to kill tumor cells highly selectively by simultaneously exploiting EpCAM as a tumor-specific cell surface molecule as well as Ras as an intracellular oncotarget in a 3D system mimicking the natural tumor microenvironment.
Collapse
Affiliation(s)
- Valentina Palacio-Castañeda
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, the Netherlands
| | - Bas van de Crommert
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, the Netherlands
| | - Elke Verploegen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, the Netherlands
| | - Mike Overeem
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, the Netherlands
| | - Jenny van Oostrum
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, the Netherlands
| | - Wouter P.R. Verdurmen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, the Netherlands
| |
Collapse
|
15
|
Yu X, Negron C, Huang L, Veldman G. TransMHCII: a novel MHC-II binding prediction model built using a protein language model and an image classifier. Antib Ther 2023; 6:137-146. [PMID: 37342671 PMCID: PMC10278228 DOI: 10.1093/abt/tbad011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/18/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
The emergence of deep learning models such as AlphaFold2 has revolutionized the structure prediction of proteins. Nevertheless, much remains unexplored, especially on how we utilize structure models to predict biological properties. Herein, we present a method using features extracted from protein language models (PLMs) to predict the major histocompatibility complex class II (MHC-II) binding affinity of peptides. Specifically, we evaluated a novel transfer learning approach where the backbone of our model was interchanged with architectures designed for image classification tasks. Features extracted from several PLMs (ESM1b, ProtXLNet or ProtT5-XL-UniRef) were passed into image models (EfficientNet v2b0, EfficientNet v2m or ViT-16). The optimal pairing of the PLM and image classifier resulted in the final model TransMHCII, outperforming NetMHCIIpan 3.2 and NetMHCIIpan 4.0-BA on the receiver operating characteristic area under the curve, balanced accuracy and Jaccard scores. The architecture innovation may facilitate the development of other deep learning models for biological problems.
Collapse
Affiliation(s)
- Xin Yu
- Biotherapeutics Discovery, AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Christopher Negron
- Biotherapeutics Discovery, AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Lili Huang
- Biotherapeutics Discovery, AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Geertruida Veldman
- Biotherapeutics Discovery, AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| |
Collapse
|
16
|
Gladilina YA, Shishparenok AN, Zhdanov DD. [Approaches for improving L-asparaginase expression in heterologous systems]. BIOMEDITSINSKAIA KHIMIIA 2023; 69:19-38. [PMID: 36857424 DOI: 10.18097/pbmc20236901019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
L-asparaginase (EC 3.5.1.1) is one of the most demanded enzymes used in the pharmaceutical industry as a drug and in the food industry to prevent the formation of toxic acrylamide. Researchers aimed to improve specific activity and reduce side effects to create safer and more potent enzyme products. However, protein modifications and heterologous expression remain problematic in the production of asparaginases from different species. Heterologous expression in optimized producer strains is rationally organized; therefore, modified and heterologous protein expression is enhanced, which is the main strategy in the production of asparaginase. This strategy solves several problems: incorrect protein folding, metabolic load on the producer strain and codon misreading, which affects translation and final protein domains, leading to a decrease in catalytic activity. The main approaches developed to improve the heterologous expression of L-asparaginases are considered in this paper.
Collapse
Affiliation(s)
| | | | - D D Zhdanov
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
17
|
Jatzlau J, Burdzinski W, Trumpp M, Obendorf L, Roßmann K, Ravn K, Hyvönen M, Bottanelli F, Broichhagen J, Knaus P. A versatile Halo- and SNAP-tagged BMP/TGFβ receptor library for quantification of cell surface ligand binding. Commun Biol 2023; 6:34. [PMID: 36635368 PMCID: PMC9837045 DOI: 10.1038/s42003-022-04388-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 12/20/2022] [Indexed: 01/14/2023] Open
Abstract
TGFβs, BMPs and Activins regulate numerous developmental and homeostatic processes and signal through hetero-tetrameric receptor complexes composed of two types of serine/threonine kinase receptors. Each of the 33 different ligands possesses unique affinities towards specific receptor types. However, the lack of specific tools hampered simultaneous testing of ligand binding towards all BMP/TGFβ receptors. Here we present a N-terminally Halo- and SNAP-tagged TGFβ/BMP receptor library to visualize receptor complexes in dual color. In combination with fluorescently labeled ligands, we established a Ligand Surface Binding Assay (LSBA) for optical quantification of receptor-dependent ligand binding in a cellular context. We highlight that LSBA is generally applicable to test (i) binding of different ligands such as Activin A, TGFβ1 and BMP9, (ii) for mutant screens and (iii) evolutionary comparisons. This experimental set-up opens opportunities for visualizing ligand-receptor binding dynamics, essential to determine signaling specificity and is easily adaptable for other receptor signaling pathways.
Collapse
Affiliation(s)
- Jerome Jatzlau
- Institute of Chemistry and Biochemistry - Biochemistry, Berlin, Germany
| | - Wiktor Burdzinski
- Institute of Chemistry and Biochemistry - Biochemistry, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), Berlin, Germany
| | - Michael Trumpp
- Institute of Chemistry and Biochemistry - Biochemistry, Berlin, Germany
| | - Leon Obendorf
- Institute of Chemistry and Biochemistry - Biochemistry, Berlin, Germany
| | - Kilian Roßmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Katharina Ravn
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | - Petra Knaus
- Institute of Chemistry and Biochemistry - Biochemistry, Berlin, Germany.
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), Berlin, Germany.
| |
Collapse
|
18
|
Liang S, Zhang C. PITHA: A Webtool to Predict Immunogenicity for Humanized and Fully Human Therapeutic Antibodies. Methods Mol Biol 2023; 2552:143-150. [PMID: 36346590 DOI: 10.1007/978-1-0716-2609-2_7] [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] [Indexed: 06/16/2023]
Abstract
Immunogenicity is an important concern to therapeutic antibodies during antibody design and development. Based on the co-crystal structures of idiotypic antibodies and their antibodies, one can see that anti-idiotypic antibodies usually bind the complementarity-determining regions (CDR) of idiotypic antibodies. Sequence and structural features, such as cavity volume at the CDR region and hydrophobicity of CDR-H3 loop region, were identified for distinguishing immunogenic antibodies from non-immunogenic antibodies. These features were integrated together with a machine learning platform to predict immunogenicity for humanized and fully human therapeutic antibodies (PITHA). This method achieved an accuracy of 83% in a leave-one-out experiment for 29 therapeutic antibodies with available crystal structures. The web server of this method is accessible at http://mabmedicine.com/PITHA or http://sysbio.unl.edu/PITHA . This method, as a step of computer-aided antibody design, helps evaluate the safety of new therapeutic antibody, which can save time and money during the therapeutic antibody development.
Collapse
Affiliation(s)
- Shide Liang
- Department of Research and Development, Bio-Thera Solutions, Guangzhou, P. R. China.
| | - Chi Zhang
- School of Biological Sciences, University of Nebraska, Lincoln, NE, USA.
| |
Collapse
|
19
|
Sulea T, Deprez C, Corbeil CR, Purisima EO. Optimizing Antibody-Antigen Binding Affinities with the ADAPT Platform. Methods Mol Biol 2023; 2552:361-374. [PMID: 36346603 DOI: 10.1007/978-1-0716-2609-2_20] [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] [Indexed: 06/16/2023]
Abstract
The ADAPT (Assisted Design of Antibody and Protein Therapeutics) platform guides the selection of mutants that improve/modulate the affinity of antibodies and other biologics. Predicted affinities are based on a consensus z-score from three scoring functions. Computational predictions are interleaved with experimental validation, significantly enhancing the robustness of the design and selection of mutants. A key step is an initial exhaustive virtual single-mutant scan that identifies hot spots and the mutations predicted to improve affinity. A small number of proposed single mutants are then produced and assayed. Only the validated single mutants (i.e., having improved affinity) are used to design double and higher-order mutants in subsequent rounds of design, avoiding the combinatorial explosion that arises from random mutagenesis. Typically, with a total of about 30-50 designed single, double, and triple mutants, affinity improvements of 10- to 100-fold are obtained.
Collapse
Affiliation(s)
- Traian Sulea
- National Research Council Canada, Human Health Therapeutics Research Centre, Montreal, QC, Canada
| | - Christophe Deprez
- National Research Council Canada, Human Health Therapeutics Research Centre, Montreal, QC, Canada
| | - Christopher R Corbeil
- National Research Council Canada, Human Health Therapeutics Research Centre, Montreal, QC, Canada
| | - Enrico O Purisima
- National Research Council Canada, Human Health Therapeutics Research Centre, Montreal, QC, Canada.
| |
Collapse
|
20
|
Charles T, Moss DL, Bhat P, Moore PW, Kummer NA, Bhattacharya A, Landry SJ, Mettu RR. CD4+ T-Cell Epitope Prediction by Combined Analysis of Antigen Conformational Flexibility and Peptide-MHCII Binding Affinity. Biochemistry 2022; 61:1585-1599. [PMID: 35834502 PMCID: PMC9352311 DOI: 10.1021/acs.biochem.2c00237] [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] [Indexed: 11/29/2022]
Abstract
![]()
Antigen processing in the class II MHC pathway depends
on conventional
proteolytic enzymes, potentially acting on antigens in native-like
conformational states. CD4+ epitope dominance arises from a competition
among antigen folding, proteolysis, and MHCII binding. Protease-sensitive
sites, linear antibody epitopes, and CD4+ T-cell epitopes were mapped
in plague vaccine candidate F1-V to evaluate the various contributions
to CD4+ epitope dominance. Using X-ray crystal structures, antigen
processing likelihood (APL) predicts CD4+ epitopes with significant
accuracy for F1-V without considering peptide-MHCII binding affinity.
We also show that APL achieves excellent performance over two benchmark
antigen sets. The profiles of conformational flexibility derived from
the X-ray crystal structures of the F1-V proteins, Caf1 and LcrV,
were similar to the biochemical profiles of linear antibody epitope
reactivity and protease sensitivity, suggesting that the role of structure
in proteolysis was captured by the analysis of the crystal structures.
The patterns of CD4+ T-cell epitope dominance in C57BL/6, CBA, and
BALB/c mice were compared to epitope predictions based on APL, MHCII
binding, or both. For a sample of 13 diverse antigens, the accuracy
of epitope prediction by the combination of APL and I-Ab-MHCII-peptide affinity reached 36%. When MHCII allele specificity
was also diverse, such as in human immunity, prediction of dominant
epitopes by APL alone reached 42% when using a stringent scoring threshold.
Because dominant CD4+ epitopes tend to occur in conformationally stable
antigen domains, crystal structures typically are available for analysis
by APL, and thus, the requirement for a crystal structure is not a
severe limitation.
Collapse
Affiliation(s)
- Tysheena Charles
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Daniel L Moss
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Pawan Bhat
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Peyton W Moore
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Nicholas A Kummer
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Avik Bhattacharya
- Department of Computer Science, Tulane University, New Orleans, Louisiana 70118, United States
| | - Samuel J Landry
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Ramgopal R Mettu
- Department of Computer Science, Tulane University, New Orleans, Louisiana 70118, United States
| |
Collapse
|
21
|
Magi Meconi G, Sasselli IR, Bianco V, Onuchic JN, Coluzza I. Key aspects of the past 30 years of protein design. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2022; 85:086601. [PMID: 35704983 DOI: 10.1088/1361-6633/ac78ef] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 06/15/2022] [Indexed: 06/15/2023]
Abstract
Proteins are the workhorse of life. They are the building infrastructure of living systems; they are the most efficient molecular machines known, and their enzymatic activity is still unmatched in versatility by any artificial system. Perhaps proteins' most remarkable feature is their modularity. The large amount of information required to specify each protein's function is analogically encoded with an alphabet of just ∼20 letters. The protein folding problem is how to encode all such information in a sequence of 20 letters. In this review, we go through the last 30 years of research to summarize the state of the art and highlight some applications related to fundamental problems of protein evolution.
Collapse
Affiliation(s)
- Giulia Magi Meconi
- Computational Biophysics Lab, Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 182, 20014, Donostia-San Sebastián, Spain
| | - Ivan R Sasselli
- Computational Biophysics Lab, Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 182, 20014, Donostia-San Sebastián, Spain
| | | | - Jose N Onuchic
- Center for Theoretical Biological Physics, Department of Physics & Astronomy, Department of Chemistry, Department of Biosciences, Rice University, Houston, TX 77251, United States of America
| | - Ivan Coluzza
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, Bld. Martina Casiano, UPV/EHU Science Park, Barrio Sarriena s/n, 48940 Leioa, Spain
- Basque Foundation for Science, Ikerbasque, 48009, Bilbao, Spain
| |
Collapse
|
22
|
Ochoa R, Lunardelli VAS, Rosa DS, Laio A, Cossio P. Multiple-Allele MHC Class II Epitope Engineering by a Molecular Dynamics-Based Evolution Protocol. Front Immunol 2022; 13:862851. [PMID: 35572587 PMCID: PMC9094701 DOI: 10.3389/fimmu.2022.862851] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Epitopes that bind simultaneously to all human alleles of Major Histocompatibility Complex class II (MHC II) are considered one of the key factors for the development of improved vaccines and cancer immunotherapies. To engineer MHC II multiple-allele binders, we developed a protocol called PanMHC-PARCE, based on the unsupervised optimization of the epitope sequence by single-point mutations, parallel explicit-solvent molecular dynamics simulations and scoring of the MHC II-epitope complexes. The key idea is accepting mutations that not only improve the affinity but also reduce the affinity gap between the alleles. We applied this methodology to enhance a Plasmodium vivax epitope for multiple-allele binding. In vitro rate-binding assays showed that four engineered peptides were able to bind with improved affinity toward multiple human MHC II alleles. Moreover, we demonstrated that mice immunized with the peptides exhibited interferon-gamma cellular immune response. Overall, the method enables the engineering of peptides with improved binding properties that can be used for the generation of new immunotherapies.
Collapse
Affiliation(s)
- Rodrigo Ochoa
- Biophysics of Tropical Diseases, Max Planck Tandem Group, University of Antioquia UdeA, Medellin, Colombia
| | | | - Daniela Santoro Rosa
- Department of Microbiology, Immunology and Parasitology, Federal University of Sao Paulo, Sao Paulo, Brazil.,Institute for Investigation in Immunology (iii), Instituto Nacional de Ciência e Tecnologia (INCT), Sao Paulo, Brazil
| | - Alessandro Laio
- Physics Area, International School for Advanced Studies (SISSA), Trieste, Italy.,Condensed Matter and Statistical Physics Section, International Centre for Theoretical Physics (ICTP), Trieste, Italy
| | - Pilar Cossio
- Biophysics of Tropical Diseases, Max Planck Tandem Group, University of Antioquia UdeA, Medellin, Colombia.,Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany.,Center for Computational Mathematics, Flatiron Institute, New York, NY, United States.,Center for Computational Biology, Flatiron Institute, New York, NY, United States
| |
Collapse
|
23
|
Tian S, Liu Y, Appleton E, Wang H, Church GM, Dong M. Targeted intracellular delivery of Cas13 and Cas9 nucleases using bacterial toxin-based platforms. Cell Rep 2022; 38:110476. [PMID: 35263584 PMCID: PMC8958846 DOI: 10.1016/j.celrep.2022.110476] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 12/26/2021] [Accepted: 02/11/2022] [Indexed: 02/06/2023] Open
Abstract
Targeted delivery of therapeutic proteins toward specific cells and across cell membranes remains major challenges. Here, we develop protein-based delivery systems utilizing detoxified single-chain bacterial toxins such as diphtheria toxin (DT) and botulinum neurotoxin (BoNT)-like toxin, BoNT/X, as carriers. The system can deliver large protein cargoes including Cas13a, CasRx, Cas9, and Cre recombinase into cells in a receptor-dependent manner, although delivery of ribonucleoproteins containing guide RNAs is not successful. Delivery of Cas13a and CasRx, together with guide RNA expression, reduces mRNAs encoding GFP, SARS-CoV-2 fragments, and endogenous proteins PPIB, KRAS, and CXCR4 in multiple cell lines. Delivery of Cre recombinase modifies the reporter loci in cells. Delivery of Cas9, together with guide RNA expression, generates mutations at the targeted genomic sites in cell lines and induced pluripotent stem cell (iPSC)-derived human neurons. These findings establish modular delivery systems based on single-chain bacterial toxins for delivery of membrane-impermeable therapeutics into targeted cells.
Collapse
Affiliation(s)
- Songhai Tian
- Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA 02115, USA.
| | - Yang Liu
- Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA 02115, USA; Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Evan Appleton
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Huan Wang
- Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - George M Church
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Min Dong
- Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
24
|
ElGamacy M. Accelerating therapeutic protein design. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 130:85-118. [PMID: 35534117 DOI: 10.1016/bs.apcsb.2022.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Protein structures provide for defined microenvironments that can support complex pharmacological functions, otherwise unachievable by small molecules. The advent of therapeutic proteins has thus greatly broadened the range of manageable disorders. Leveraging the knowledge and recent advances in de novo protein design methods has the prospect of revolutionizing how protein drugs are discovered and developed. This review lays out the main challenges facing therapeutic proteins discovery and development, and how present and future advancements of protein design can accelerate the protein drug pipelines.
Collapse
Affiliation(s)
- Mohammad ElGamacy
- University Hospital Tübingen, Division of Translational Oncology, Tübingen, Germany; Max Planck Institute for Biology, Tübingen, Germany.
| |
Collapse
|
25
|
Mould RC, van Vloten JP, AuYeung AWK, Walsh SR, de Jong J, Susta L, Mutsaers AJ, Petrik JJ, Wood GA, Wootton SK, Karimi K, Bridle BW. Using a Prime-Boost Vaccination Strategy That Proved Effective for High Resolution Epitope Mapping to Characterize the Elusive Immunogenicity of Survivin. Cancers (Basel) 2021; 13:cancers13246270. [PMID: 34944889 PMCID: PMC8699342 DOI: 10.3390/cancers13246270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The generation of tumor-specific T cells remains a pillar of modern cancer immunotherapy. Exogenous vaccines often rely on targeting tumor-associated antigens. The anti-apoptotic protein survivin has been deemed a high priority target due to its overexpression in a wide variety of tumor types. To support the analysis of tumor-associated T cell responses, optimization of epitope mapping would be valuable. A heterologous prime-boost vaccination strategy was designed to target survivin to induce anti-tumor immune responses. However, survivin-specific T cell responses could not be detected in mice. Potential mechanisms to explain this failure were explored. To confirm the robustness of the vaccination platform, enhanced green fluorescent protein (eGFP) was targeted since it has been defined as a protein with relatively low immunogenicity. In this context the vaccination strategy uncovered novel T cell epitopes from eGFP in two strains of mice. This research highlighted the utility of the vaccine platform to triage potential target antigens based on their immunogenicity. Abstract Survivin is a member of the inhibitor of apoptosis family of proteins and has been reported to be highly expressed in a variety of cancer types, making it a high priority target for cancer vaccination. We previously described a heterologous prime-boost strategy using a replication-deficient adenovirus, followed by an oncolytic rhabdovirus that generates unprecedented antigen-specific T cell responses. We engineered each vector to express a mutated version of full-length murine survivin. We first sought to uncover the complete epitope map for survivin-specific T cell responses in C57BL/6 and BALB/c mice by flow cytometry. However, no T cell responses were detected by intracellular cytokine staining after re-stimulation of T cells. Survivin has been found to be expressed by activated T cells, which could theoretically cause T cell-mediated killing of activated T cells, known as fratricide. We were unable to recapitulate this phenomenon in experiments. Interestingly, the inactivated survivin construct has been previously shown to directly kill tumor cells in vitro. However, there was no evidence in our models of induction of death in antigen-presenting cells due to treatment with a survivin-expressing vector. Using the same recombinant virus-vectored prime-boost strategy targeting the poorly immunogenic enhanced green fluorescent protein proved to be a highly sensitive method for mapping T cell epitopes, particularly in the context of identifying novel epitopes recognized by CD4+ T cells. Overall, these results suggested there may be unusually robust tolerance to survivin in commonly used mouse strains that cannot be broken, even when using a particularly potent vaccination platform. However, the vaccination method shows great promise as a strategy for identifying novel and subdominant T cell epitopes.
Collapse
Affiliation(s)
- Robert C. Mould
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.C.M.); (J.P.v.V.); (A.W.K.A.); (J.d.J.); (L.S.); (G.A.W.); (S.K.W.); (K.K.)
| | - Jacob P. van Vloten
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.C.M.); (J.P.v.V.); (A.W.K.A.); (J.d.J.); (L.S.); (G.A.W.); (S.K.W.); (K.K.)
| | - Amanda W. K. AuYeung
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.C.M.); (J.P.v.V.); (A.W.K.A.); (J.d.J.); (L.S.); (G.A.W.); (S.K.W.); (K.K.)
| | - Scott R. Walsh
- McMaster Immunology Research Centre, McMaster University Hamilton, Hamilton, ON L8S 3L8, Canada;
| | - Jondavid de Jong
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.C.M.); (J.P.v.V.); (A.W.K.A.); (J.d.J.); (L.S.); (G.A.W.); (S.K.W.); (K.K.)
| | - Leonardo Susta
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.C.M.); (J.P.v.V.); (A.W.K.A.); (J.d.J.); (L.S.); (G.A.W.); (S.K.W.); (K.K.)
| | - Anthony J. Mutsaers
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.J.M.); (J.J.P.)
| | - James J. Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.J.M.); (J.J.P.)
| | - Geoffrey A. Wood
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.C.M.); (J.P.v.V.); (A.W.K.A.); (J.d.J.); (L.S.); (G.A.W.); (S.K.W.); (K.K.)
| | - Sarah K. Wootton
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.C.M.); (J.P.v.V.); (A.W.K.A.); (J.d.J.); (L.S.); (G.A.W.); (S.K.W.); (K.K.)
| | - Khalil Karimi
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.C.M.); (J.P.v.V.); (A.W.K.A.); (J.d.J.); (L.S.); (G.A.W.); (S.K.W.); (K.K.)
| | - Byram W. Bridle
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.C.M.); (J.P.v.V.); (A.W.K.A.); (J.d.J.); (L.S.); (G.A.W.); (S.K.W.); (K.K.)
- Correspondence: ; Tel.: +51-9824-4120 (ext. 54657)
| |
Collapse
|
26
|
Chen BM, Cheng TL, Roffler SR. Polyethylene Glycol Immunogenicity: Theoretical, Clinical, and Practical Aspects of Anti-Polyethylene Glycol Antibodies. ACS NANO 2021; 15:14022-14048. [PMID: 34469112 DOI: 10.1021/acsnano.1c05922] [Citation(s) in RCA: 260] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Polyethylene glycol (PEG) is a flexible, hydrophilic simple polymer that is physically attached to peptides, proteins, nucleic acids, liposomes, and nanoparticles to reduce renal clearance, block antibody and protein binding sites, and enhance the half-life and efficacy of therapeutic molecules. Some naïve individuals have pre-existing antibodies that can bind to PEG, and some PEG-modified compounds induce additional antibodies against PEG, which can adversely impact drug efficacy and safety. Here we provide a framework to better understand PEG immunogenicity and how antibodies against PEG affect pegylated drug and nanoparticles. Analysis of published studies reveals rules for predicting accelerated blood clearance of pegylated medicine and therapeutic liposomes. Experimental studies of anti-PEG antibody binding to different forms, sizes, and immobilization states of PEG are also provided. The widespread use of SARS-CoV-2 RNA vaccines that incorporate PEG in lipid nanoparticles make understanding possible effects of anti-PEG antibodies on pegylated medicines even more critical.
Collapse
Affiliation(s)
- Bing-Mae Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Tian-Lu Cheng
- Center for Biomarkers and Biotech Drugs, Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
27
|
Guaranteed Diversity and Optimality in Cost Function Network Based Computational Protein Design Methods. ALGORITHMS 2021. [DOI: 10.3390/a14060168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Proteins are the main active molecules of life. Although natural proteins play many roles, as enzymes or antibodies for example, there is a need to go beyond the repertoire of natural proteins to produce engineered proteins that precisely meet application requirements, in terms of function, stability, activity or other protein capacities. Computational Protein Design aims at designing new proteins from first principles, using full-atom molecular models. However, the size and complexity of proteins require approximations to make them amenable to energetic optimization queries. These approximations make the design process less reliable, and a provable optimal solution may fail. In practice, expensive libraries of solutions are therefore generated and tested. In this paper, we explore the idea of generating libraries of provably diverse low-energy solutions by extending cost function network algorithms with dedicated automaton-based diversity constraints on a large set of realistic full protein redesign problems. We observe that it is possible to generate provably diverse libraries in reasonable time and that the produced libraries do enhance the Native Sequence Recovery, a traditional measure of design methods reliability.
Collapse
|
28
|
Yachnin BJ, Mulligan VK, Khare SD, Bailey-Kellogg C. MHCEpitopeEnergy, a Flexible Rosetta-Based Biotherapeutic Deimmunization Platform. J Chem Inf Model 2021; 61:2368-2382. [PMID: 33900750 PMCID: PMC8225355 DOI: 10.1021/acs.jcim.1c00056] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
As non-"self" macromolecules, biotherapeutics can trigger an immune response that can reduce drug efficacy, require patients to be taken off therapy, or even cause life-threatening reactions. To enable the flexible and facile design of protein biotherapeutics while reducing the prevalence of T-cell epitopes that drive immune recognition, we have integrated into the Rosetta protein design suite a new scoring term that allows design protocols to account for predicted or experimentally identified epitopes in the optimized objective function. This flexible scoring term can be used in any Rosetta design trajectory, can be targeted to specific regions of a protein, and can be readily extended to work with a variety of epitope predictors. By performing extensive design runs with varied design parameter choices for three case study proteins as well as a larger diverse benchmark, we show that the incorporation of this scoring term enables the effective exploration of an alternative, deimmunized sequence space to discover diverse proteins that are potentially highly deimmunized while retaining physical and chemical qualities similar to those yielded by equivalent nondeimmunizing sequence design protocols.
Collapse
Affiliation(s)
- Brahm J. Yachnin
- Department of Chemistry and Chemical Biology and Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Vikram Khipple Mulligan
- Center for Computational Biology, Flatiron Institute, 162 Fifth Avenue, New York, NY, 10010, USA
| | - Sagar D. Khare
- Department of Chemistry and Chemical Biology and Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | | |
Collapse
|
29
|
Gu C, Cao X, Wang Z, Hu X, Yao Y, Zhou Y, Liu P, Liu X, Gao G, Hu X, Zhang Y, Chen Z, Gao L, Peng Y, Jia F, Shan C, Yu L, Liu K, Li N, Guo W, Jiang G, Min J, Zhang J, Yang L, Shi M, Hou T, Li Y, Liang W, Lu G, Yang C, Wang Y, Xia K, Xiao Z, Xue J, Huang X, Chen X, Ma H, Song D, Pan Z, Wang X, Guo H, Liang H, Yuan Z, Guan W, Deng SJ. A human antibody of potent efficacy against SARS-CoV-2 in rhesus macaques showed strong blocking activity to B.1.351. MAbs 2021; 13:1930636. [PMID: 34097570 PMCID: PMC8189090 DOI: 10.1080/19420862.2021.1930636] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 12/25/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), which causes coronavirus disease-2019 (COVID-19), interacts with the host cell receptor angiotensin-converting enzyme 2 (hACE2) via its spike 1 protein during infection. After the virus sequence was published, we identified two potent antibodies against the SARS-CoV-2 receptor binding domain (RBD) from antibody libraries using a phage-to-yeast (PtY) display platform in only 10 days. Our lead antibody JMB2002, now in a Phase 1 clinical trial (ChiCTR2100042150), showed broad-spectrum in vitro blocking activity against hACE2 binding to the RBD of multiple SARS-CoV-2 variants, including B.1.351 that was reportedly much more resistant to neutralization by convalescent plasma, vaccine sera and some clinical-stage neutralizing antibodies. Furthermore, JMB2002 has demonstrated complete prophylactic and potent therapeutic efficacy in a rhesus macaque disease model. Prophylactic and therapeutic countermeasure intervention of SARS-CoV-2 using JMB2002 would likely slow down the transmission of currently emerged SARS-CoV-2 variants and result in more efficient control of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Chunyin Gu
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Xiaodan Cao
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Zongda Wang
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Xue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| | - Yanfeng Yao
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, People’s Republic of China
| | - Yiwu Zhou
- Department of Forensic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Peipei Liu
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Xiaowu Liu
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Ge Gao
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, People’s Republic of China
| | - Xiao Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| | - Yecheng Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| | - Zhen Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| | - Li Gao
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Yun Peng
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, People’s Republic of China
| | - Fangfang Jia
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Chao Shan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| | - Li Yu
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Kunpeng Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| | - Nan Li
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Weiwei Guo
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| | - Guoping Jiang
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Juan Min
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, People’s Republic of China
| | - Jianjian Zhang
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Lu Yang
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Meng Shi
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Tianquan Hou
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Yanan Li
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Weichen Liang
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Guoqiao Lu
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Congyi Yang
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Yuting Wang
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Kaiwen Xia
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Zheng Xiao
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Jianhua Xue
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Xueyi Huang
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Xin Chen
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Haixia Ma
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, People’s Republic of China
| | - Donglin Song
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, People’s Republic of China
| | - Zhongzong Pan
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Xueping Wang
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Haibing Guo
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Hong Liang
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| | - Zhiming Yuan
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, People’s Republic of China
| | - Wuxiang Guan
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, People’s Republic of China
| | - Su-Jun Deng
- Shanghai Jemincare Pharmaceuticals Co., Ltd., Shanghai, People’s Republic of China
| |
Collapse
|
30
|
Ong E, Huang X, Pearce R, Zhang Y, He Y. Computational design of SARS-CoV-2 spike glycoproteins to increase immunogenicity by T cell epitope engineering. Comput Struct Biotechnol J 2020; 19:518-529. [PMID: 33398234 PMCID: PMC7773544 DOI: 10.1016/j.csbj.2020.12.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/24/2020] [Accepted: 12/24/2020] [Indexed: 01/12/2023] Open
Abstract
The development of effective and safe vaccines is the ultimate way to efficiently stop the ongoing COVID-19 pandemic, which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Built on the fact that SARS-CoV-2 utilizes the association of its Spike (S) protein with the human angiotensin-converting enzyme 2 (ACE2) receptor to invade host cells, we computationally redesigned the S protein sequence to improve its immunogenicity and antigenicity. Toward this purpose, we extended an evolutionary protein design algorithm, EvoDesign, to create thousands of stable S protein variants that perturb the core protein sequence but keep the surface conformation and B cell epitopes. The T cell epitope content and similarity scores of the perturbed sequences were calculated and evaluated. Out of 22,914 designs with favorable stability energy, 301 candidates contained at least two pre-existing immunity-related epitopes and had promising immunogenic potential. The benchmark tests showed that, although the epitope restraints were not included in the scoring function of EvoDesign, the top S protein design successfully recovered 31 out of the 32 major histocompatibility complex (MHC)-II T cell promiscuous epitopes in the native S protein, where two epitopes were present in all seven human coronaviruses. Moreover, the newly designed S protein introduced nine new MHC-II T cell promiscuous epitopes that do not exist in the wildtype SARS-CoV-2. These results demonstrated a new and effective avenue to enhance a target protein's immunogenicity using rational protein design, which could be applied for new vaccine design against COVID-19 and other pathogens.
Collapse
Affiliation(s)
- Edison Ong
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiaoqiang Huang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Robin Pearce
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yang Zhang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yongqun He
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
31
|
Zinsli LV, Stierlin N, Loessner MJ, Schmelcher M. Deimmunization of protein therapeutics - Recent advances in experimental and computational epitope prediction and deletion. Comput Struct Biotechnol J 2020; 19:315-329. [PMID: 33425259 PMCID: PMC7779837 DOI: 10.1016/j.csbj.2020.12.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022] Open
Abstract
Biotherapeutics, and antimicrobial proteins in particular, are of increasing interest for human medicine. An important challenge in the development of such therapeutics is their potential immunogenicity, which can induce production of anti-drug-antibodies, resulting in altered pharmacokinetics, reduced efficacy, and potentially severe anaphylactic or hypersensitivity reactions. For this reason, the development and application of effective deimmunization methods for protein drugs is of utmost importance. Deimmunization may be achieved by unspecific shielding approaches, which include PEGylation, fusion to polypeptides (e.g., XTEN or PAS), reductive methylation, glycosylation, and polysialylation. Alternatively, the identification of epitopes for T cells or B cells and their subsequent deletion through site-directed mutagenesis represent promising deimmunization strategies and can be accomplished through either experimental or computational approaches. This review highlights the most recent advances and current challenges in the deimmunization of protein therapeutics, with a special focus on computational epitope prediction and deletion tools.
Collapse
Key Words
- ABR, Antigen-binding region
- ADA, Anti-drug antibody
- ANN, Artificial neural network
- APC, Antigen-presenting cell
- Anti-drug-antibody
- B cell epitope
- BCR, B cell receptor
- Bab, Binding antibody
- CDR, Complementarity determining region
- CRISPR, Clustered regularly interspaced short palindromic repeats
- DC, Dendritic cell
- ELP, Elastin-like polypeptide
- EPO, Erythropoietin
- ER, Endoplasmatic reticulum
- GLK, Gelatin-like protein
- HAP, Homo-amino-acid polymer
- HLA, Human leukocyte antigen
- HMM, Hidden Markov model
- IL, Interleukin
- Ig, Immunoglobulin
- Immunogenicity
- LPS, Lipopolysaccharide
- MHC, Major histocompatibility complex
- NMR, Nuclear magnetic resonance
- Nab, Neutralizing antibody
- PAMP, Pathogen-associated molecular pattern
- PAS, Polypeptide composed of proline, alanine, and/or serine
- PBMC, Peripheral blood mononuclear cell
- PD, Pharmacodynamics
- PEG, Polyethylene glycol
- PK, Pharmacokinetics
- PRR, Pattern recognition receptor
- PSA, Sialic acid polymers
- Protein therapeutic
- RNN, Recurrent artificial neural network
- SVM, Support vector machine
- T cell epitope
- TAP, Transporter associated with antigen processing
- TCR, T cell receptor
- TLR, Toll-like receptor
- XTEN, “Xtended” recombinant polypeptide
Collapse
Affiliation(s)
- Léa V. Zinsli
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Noël Stierlin
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Martin J. Loessner
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Mathias Schmelcher
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Khramtsov YV, Vlasova AD, Vlasov AV, Rosenkranz AA, Ulasov AV, Ryzhykau YL, Kuklin AI, Orekhov AS, Eydlin IB, Georgiev GP, Gordeliy VI, Sobolev AS. Low-resolution structures of modular nanotransporters shed light on their functional activity. ACTA CRYSTALLOGRAPHICA SECTION D-STRUCTURAL BIOLOGY 2020; 76:1270-1279. [PMID: 33263332 DOI: 10.1107/s2059798320013765] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022]
Abstract
Modular nanotransporters (MNTs) are multifunctional chimeric polypeptides for the multistep transport of locally acting cytotoxic agents into the nuclei of cancer target cells. MNTs consist of several polypeptide domains (functional modules) for the recognition of a cell-surface internalizable receptor, pH-dependent endosomal escape and subsequent transport into the nucleus through the nuclear pores. MNTs are a promising means for cancer treatment. As has been shown previously, all of the modules of MNTs retain their functionalities. Despite their importance, there is no structural information available about these chimeric polypeptides, which hampers the creation of new MNT variants. Here, a low-resolution 3D structure of an MNT is presented which was obtained by atomic force microscopy, transmission electron microscopy and small-angle X-ray scattering coupled to size-exclusion chromatography. The data suggest that the MNT can adopt two main conformations, but in both conformations the protein N- and C-termini are distanced and do not influence each other. The change in the MNT conformation during acidification of the medium was also studied. It was shown that the fraction of the elongated conformation increases upon acidification. The results of this work will be useful for the development of MNTs that are suitable for clinical trials and possible therapeutic applications.
Collapse
Affiliation(s)
- Yuri V Khramtsov
- Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russian Federation
| | - Anastasiia D Vlasova
- Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russian Federation
| | - Alexey V Vlasov
- Moscow Institute of Physics and Technology, Dolgoprudny 141700, Russian Federation
| | - Andrey A Rosenkranz
- Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russian Federation
| | - Alexey V Ulasov
- Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russian Federation
| | - Yury L Ryzhykau
- Moscow Institute of Physics and Technology, Dolgoprudny 141700, Russian Federation
| | - Alexander I Kuklin
- Moscow Institute of Physics and Technology, Dolgoprudny 141700, Russian Federation
| | - Anton S Orekhov
- Moscow Institute of Physics and Technology, Dolgoprudny 141700, Russian Federation
| | - Ilia B Eydlin
- Moscow Institute of Physics and Technology, Dolgoprudny 141700, Russian Federation
| | - Georgii P Georgiev
- Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russian Federation
| | - Valentin I Gordeliy
- Moscow Institute of Physics and Technology, Dolgoprudny 141700, Russian Federation
| | - Alexander S Sobolev
- Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russian Federation
| |
Collapse
|
33
|
Nerli S, Sgourakis NG. Structure-Based Modeling of SARS-CoV-2 Peptide/HLA-A02 Antigens. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:553478. [PMID: 35047875 PMCID: PMC8757863 DOI: 10.3389/fmedt.2020.553478] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 10/07/2020] [Indexed: 11/13/2022] Open
Abstract
SARS-CoV-2-specific CD4 and CD8 T cells have been shown to be present in individuals with acute, mild, and asymptomatic Coronavirus disease (COVID-19). Toward the development of diagnostic and therapeutic tools to fight COVID-19, it is important to predict and characterize T cell epitopes expressed by SARS-CoV-2. Here, we use RosettaMHC, a comparative modeling approach which leverages existing structures of peptide/MHC complexes available in the Protein Data Bank, to derive accurate 3D models for putative SARS-CoV-2 CD8 epitopes. We outline an application of our method to model 8-10 residue epitopic peptides predicted to bind to the common allele HLA-A*02:01, and we make our models publicly available through an online database (https://rosettamhc.chemistry.ucsc.edu). We further compare electrostatic surfaces with models of homologous peptide/HLA-A*02:01 complexes from human common cold coronavirus strains to identify epitopes which may be recognized by a shared pool of cross-reactive TCRs. As more detailed studies on antigen-specific T cell recognition become available, RosettaMHC models can be used to understand the link between peptide/HLA complex structure and surface chemistry with immunogenicity, in the context of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Santrupti Nerli
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Nikolaos G. Sgourakis
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
34
|
Mitigation of T-cell dependent immunogenicity by reengineering factor VIIa analogue. Blood Adv 2020; 3:2668-2678. [PMID: 31506285 DOI: 10.1182/bloodadvances.2019000338] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/03/2019] [Indexed: 02/08/2023] Open
Abstract
Vatreptacog alfa (VA), a recombinant activated human factor VII (rFVIIa) variant with 3 amino acid substitutions, was developed to provide increased procoagulant activity in hemophilia patients with inhibitors to factor VIII or factor IX. In phase 3 clinical trials, changes introduced during the bioengineering of VA resulted in the development of undesired anti-drug antibodies in some patients, leading to the termination of a potentially promising therapeutic protein product. Here, we use preclinical biomarkers associated with clinical immunogenicity to validate our deimmunization strategy applied to this bioengineered rFVIIa analog. The reengineered rFVIIa analog variants retained increased intrinsic thrombin generation activity but did not elicit T-cell responses in peripheral blood mononuclear cells isolated from 50 HLA typed subjects representing the human population. Our algorithm, rational immunogenicity determination, offers a broadly applicable deimmunizing strategy for bioengineered proteins.
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
|
Zajc CU, Salzer B, Taft JM, Reddy ST, Lehner M, Traxlmayr MW. Driving CARs with alternative navigation tools - the potential of engineered binding scaffolds. FEBS J 2020; 288:2103-2118. [PMID: 32794303 PMCID: PMC8048499 DOI: 10.1111/febs.15523] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/31/2020] [Accepted: 08/08/2020] [Indexed: 12/11/2022]
Abstract
T cells that are genetically engineered to express chimeric antigen receptors (CAR T cells) have shown impressive clinical efficacy against B‐cell malignancies. In contrast to these highly potent CD19‐targeting CAR T cells, many of those directed against other tumor entities and antigens currently suffer from several limitations. For example, it has been demonstrated that many scFvs used as antigen‐binding domains in CARs show some degree of oligomerization, which leads to tonic signaling, T cell exhaustion, and poor performance in vivo. Therefore, in many cases alternatives to scFvs would be beneficial. Fortunately, due to the development of powerful protein engineering technologies, also non‐immunoglobulin‐based scaffolds can be engineered to specifically recognize antigens, thus eliminating the historical dependence on antibody‐based binding domains. Here, we discuss the advantages and disadvantages of such engineered binding scaffolds, in particular with respect to their application in CARs. We review recent studies, collectively showing that there is no functional or biochemical aspect that necessitates the use of scFvs in CARs. Instead, antigen recognition can also be mediated efficiently by engineered binding scaffolds, as well as natural ligands or receptors fused to the CAR backbone. Finally, we critically discuss the risk of immunogenicity and show that the extent of nonhuman amino acid stretches in engineered scaffolds—even in those based on nonhuman proteins—is more similar to humanized scFvs than might be anticipated. Together, we expect that engineered binding scaffolds and natural ligands and receptors will be increasingly used for the design of CAR T cells.
Collapse
Affiliation(s)
- Charlotte U Zajc
- Christian Doppler Laboratory for Next Generation CAR T Cells, Vienna, Austria.,Department of Chemistry, Institute of Biochemistry, BOKU-University of Natural Resources and Life Sciences, Vienna, Austria
| | - Benjamin Salzer
- Christian Doppler Laboratory for Next Generation CAR T Cells, Vienna, Austria.,St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Joseph M Taft
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Sai T Reddy
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Manfred Lehner
- Christian Doppler Laboratory for Next Generation CAR T Cells, Vienna, Austria.,St. Anna Children's Cancer Research Institute, Vienna, Austria.,Department of Pediatrics, St. Anna Kinderspital, Medical University of Vienna, Austria
| | - Michael W Traxlmayr
- Christian Doppler Laboratory for Next Generation CAR T Cells, Vienna, Austria.,Department of Chemistry, Institute of Biochemistry, BOKU-University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
37
|
Ong E, Huang X, Pearce R, Zhang Y, He Y. Rational Design of SARS-CoV-2 Spike Glycoproteins To Increase Immunogenicity By T Cell Epitope Engineering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.08.14.251496. [PMID: 32817949 PMCID: PMC7430581 DOI: 10.1101/2020.08.14.251496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The current COVID-19 pandemic caused by SARS-CoV-2 has resulted in millions of confirmed cases and thousands of deaths globally. Extensive efforts and progress have been made to develop effective and safe vaccines against COVID-19. A primary target of these vaccines is the SARS-CoV-2 spike (S) protein, and many studies utilized structural vaccinology techniques to either stabilize the protein or fix the receptor-binding domain at certain states. In this study, we extended an evolutionary protein design algorithm, EvoDesign, to create thousands of stable S protein variants without perturbing the surface conformation and B cell epitopes of the S protein. We then evaluated the mutated S protein candidates based on predicted MHC-II T cell promiscuous epitopes as well as the epitopes' similarity to human peptides. The presented strategy aims to improve the S protein's immunogenicity and antigenicity by inducing stronger CD4 T cell response while maintaining the protein's native structure and function. The top EvoDesign S protein candidate (Design-10705) recovered 31 out of 32 MHC-II T cell promiscuous epitopes in the native S protein, in which two epitopes were present in all seven human coronaviruses. This newly designed S protein also introduced nine new MHC-II T cell promiscuous epitopes and showed high structural similarity to its native conformation. The proposed structural vaccinology method provides an avenue to rationally design the antigen's structure with increased immunogenicity, which could be applied to the rational design of new COVID-19 vaccine candidates.
Collapse
Affiliation(s)
- Edison Ong
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiaoqiang Huang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Robin Pearce
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yang Zhang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yongqun He
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
38
|
Wollacott AM, Xue C, Qin Q, Hua J, Bohnuud T, Viswanathan K, Kolachalama VB. Quantifying the nativeness of antibody sequences using long short-term memory networks. Protein Eng Des Sel 2020; 32:347-354. [PMID: 31504835 PMCID: PMC7372931 DOI: 10.1093/protein/gzz031] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/11/2019] [Accepted: 07/07/2019] [Indexed: 11/12/2022] Open
Abstract
Antibodies often undergo substantial engineering en route to the generation of a therapeutic candidate with good developability properties. Characterization of antibody libraries has shown that retaining native-like sequence improves the overall quality of the library. Motivated by recent advances in deep learning, we developed a bi-directional long short-term memory (LSTM) network model to make use of the large amount of available antibody sequence information, and use this model to quantify the nativeness of antibody sequences. The model scores sequences for their similarity to naturally occurring antibodies, which can be used as a consideration during design and engineering of libraries. We demonstrate the performance of this approach by training a model on human antibody sequences and show that our method outperforms other approaches at distinguishing human antibodies from those of other species. We show the applicability of this method for the evaluation of synthesized antibody libraries and humanization of mouse antibodies.
Collapse
Affiliation(s)
| | - Chonghua Xue
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Qiuyuan Qin
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - June Hua
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | - Vijaya B Kolachalama
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA.,Hariri Institute of Computing and Computational Science & Engineering, Boston University, Boston, MA 02115, USA.,Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA.,Boston University Alzheimer's Disease Center, Boston, MA 02118, USA
| |
Collapse
|
39
|
Leman JK, Weitzner BD, Lewis SM, Adolf-Bryfogle J, Alam N, Alford RF, Aprahamian M, Baker D, Barlow KA, Barth P, Basanta B, Bender BJ, Blacklock K, Bonet J, Boyken SE, Bradley P, Bystroff C, Conway P, Cooper S, Correia BE, Coventry B, Das R, De Jong RM, DiMaio F, Dsilva L, Dunbrack R, Ford AS, Frenz B, Fu DY, Geniesse C, Goldschmidt L, Gowthaman R, Gray JJ, Gront D, Guffy S, Horowitz S, Huang PS, Huber T, Jacobs TM, Jeliazkov JR, Johnson DK, Kappel K, Karanicolas J, Khakzad H, Khar KR, Khare SD, Khatib F, Khramushin A, King IC, Kleffner R, Koepnick B, Kortemme T, Kuenze G, Kuhlman B, Kuroda D, Labonte JW, Lai JK, Lapidoth G, Leaver-Fay A, Lindert S, Linsky T, London N, Lubin JH, Lyskov S, Maguire J, Malmström L, Marcos E, Marcu O, Marze NA, Meiler J, Moretti R, Mulligan VK, Nerli S, Norn C, Ó'Conchúir S, Ollikainen N, Ovchinnikov S, Pacella MS, Pan X, Park H, Pavlovicz RE, Pethe M, Pierce BG, Pilla KB, Raveh B, Renfrew PD, Burman SSR, Rubenstein A, Sauer MF, Scheck A, Schief W, Schueler-Furman O, Sedan Y, Sevy AM, Sgourakis NG, Shi L, Siegel JB, Silva DA, Smith S, Song Y, et alLeman JK, Weitzner BD, Lewis SM, Adolf-Bryfogle J, Alam N, Alford RF, Aprahamian M, Baker D, Barlow KA, Barth P, Basanta B, Bender BJ, Blacklock K, Bonet J, Boyken SE, Bradley P, Bystroff C, Conway P, Cooper S, Correia BE, Coventry B, Das R, De Jong RM, DiMaio F, Dsilva L, Dunbrack R, Ford AS, Frenz B, Fu DY, Geniesse C, Goldschmidt L, Gowthaman R, Gray JJ, Gront D, Guffy S, Horowitz S, Huang PS, Huber T, Jacobs TM, Jeliazkov JR, Johnson DK, Kappel K, Karanicolas J, Khakzad H, Khar KR, Khare SD, Khatib F, Khramushin A, King IC, Kleffner R, Koepnick B, Kortemme T, Kuenze G, Kuhlman B, Kuroda D, Labonte JW, Lai JK, Lapidoth G, Leaver-Fay A, Lindert S, Linsky T, London N, Lubin JH, Lyskov S, Maguire J, Malmström L, Marcos E, Marcu O, Marze NA, Meiler J, Moretti R, Mulligan VK, Nerli S, Norn C, Ó'Conchúir S, Ollikainen N, Ovchinnikov S, Pacella MS, Pan X, Park H, Pavlovicz RE, Pethe M, Pierce BG, Pilla KB, Raveh B, Renfrew PD, Burman SSR, Rubenstein A, Sauer MF, Scheck A, Schief W, Schueler-Furman O, Sedan Y, Sevy AM, Sgourakis NG, Shi L, Siegel JB, Silva DA, Smith S, Song Y, Stein A, Szegedy M, Teets FD, Thyme SB, Wang RYR, Watkins A, Zimmerman L, Bonneau R. Macromolecular modeling and design in Rosetta: recent methods and frameworks. Nat Methods 2020; 17:665-680. [PMID: 32483333 PMCID: PMC7603796 DOI: 10.1038/s41592-020-0848-2] [Show More Authors] [Citation(s) in RCA: 494] [Impact Index Per Article: 98.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 04/22/2020] [Indexed: 12/12/2022]
Abstract
The Rosetta software for macromolecular modeling, docking and design is extensively used in laboratories worldwide. During two decades of development by a community of laboratories at more than 60 institutions, Rosetta has been continuously refactored and extended. Its advantages are its performance and interoperability between broad modeling capabilities. Here we review tools developed in the last 5 years, including over 80 methods. We discuss improvements to the score function, user interfaces and usability. Rosetta is available at http://www.rosettacommons.org.
Collapse
Affiliation(s)
- Julia Koehler Leman
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA.
- Department of Biology, New York University, New York, New York, USA.
| | - Brian D Weitzner
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Lyell Immunopharma Inc., Seattle, WA, USA
| | - Steven M Lewis
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry, Duke University, Durham, NC, USA
- Cyrus Biotechnology, Seattle, WA, USA
| | - Jared Adolf-Bryfogle
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Nawsad Alam
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rebecca F Alford
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Melanie Aprahamian
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Kyle A Barlow
- Graduate Program in Bioinformatics, University of California San Francisco, San Francisco, CA, USA
| | - Patrick Barth
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Baylor College of Medicine, Department of Pharmacology, Houston, TX, USA
| | - Benjamin Basanta
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Biological Physics Structure and Design PhD Program, University of Washington, Seattle, WA, USA
| | - Brian J Bender
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Kristin Blacklock
- Institute of Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Jaume Bonet
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Scott E Boyken
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Lyell Immunopharma Inc., Seattle, WA, USA
| | - Phil Bradley
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Chris Bystroff
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Patrick Conway
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Seth Cooper
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA
| | - Bruno E Correia
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Brian Coventry
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Rhiju Das
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Frank DiMaio
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lorna Dsilva
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA
| | - Roland Dunbrack
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Alexander S Ford
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Brandon Frenz
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Cyrus Biotechnology, Seattle, WA, USA
| | - Darwin Y Fu
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Caleb Geniesse
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Ragul Gowthaman
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Jeffrey J Gray
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD, USA
| | - Dominik Gront
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Sharon Guffy
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Scott Horowitz
- Department of Chemistry & Biochemistry, University of Denver, Denver, CO, USA
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Po-Ssu Huang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Thomas Huber
- Research School of Chemistry, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Tim M Jacobs
- Program in Bioinformatics and Computational Biology, Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - David K Johnson
- Center for Computational Biology, University of Kansas, Lawrence, KS, USA
| | - Kalli Kappel
- Biophysics Program, Stanford University, Stanford, CA, USA
| | - John Karanicolas
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Hamed Khakzad
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Institute for Computational Science, University of Zurich, Zurich, Switzerland
- S3IT, University of Zurich, Zurich, Switzerland
| | - Karen R Khar
- Cyrus Biotechnology, Seattle, WA, USA
- Center for Computational Biology, University of Kansas, Lawrence, KS, USA
| | - Sagar D Khare
- Institute of Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Department of Chemistry and Chemical Biology, The State University of New Jersey, Piscataway, NJ, USA
- Center for Integrative Proteomics Research, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Computational Biology and Molecular Biophysics Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Firas Khatib
- Department of Computer and Information Science, University of Massachusetts Dartmouth, Dartmouth, MA, USA
| | - Alisa Khramushin
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Indigo C King
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Cyrus Biotechnology, Seattle, WA, USA
| | - Robert Kleffner
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA
| | - Brian Koepnick
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Tanja Kortemme
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Georg Kuenze
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
| | - Brian Kuhlman
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daisuke Kuroda
- Medical Device Development and Regulation Research Center, School of Engineering, University of Tokyo, Tokyo, Japan
- Department of Bioengineering, School of Engineering, University of Tokyo, Tokyo, Japan
| | - Jason W Labonte
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, USA
| | - Jason K Lai
- Baylor College of Medicine, Department of Pharmacology, Houston, TX, USA
| | - Gideon Lapidoth
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Andrew Leaver-Fay
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Steffen Lindert
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, USA
| | - Thomas Linsky
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Nir London
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Joseph H Lubin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sergey Lyskov
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jack Maguire
- Program in Bioinformatics and Computational Biology, Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lars Malmström
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Institute for Computational Science, University of Zurich, Zurich, Switzerland
- S3IT, University of Zurich, Zurich, Switzerland
- Division of Infection Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Enrique Marcos
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Research in Biomedicine Barcelona, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Orly Marcu
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nicholas A Marze
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
- Departments of Chemistry, Pharmacology and Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
- Institute for Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | - Rocco Moretti
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Vikram Khipple Mulligan
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Santrupti Nerli
- Department of Computer Science, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Christoffer Norn
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Shane Ó'Conchúir
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Noah Ollikainen
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Sergey Ovchinnikov
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Michael S Pacella
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Xingjie Pan
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Hahnbeom Park
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Ryan E Pavlovicz
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Cyrus Biotechnology, Seattle, WA, USA
| | - Manasi Pethe
- Department of Chemistry and Chemical Biology, The State University of New Jersey, Piscataway, NJ, USA
- Center for Integrative Proteomics Research, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Brian G Pierce
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Kala Bharath Pilla
- Research School of Chemistry, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Barak Raveh
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - P Douglas Renfrew
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Shourya S Roy Burman
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Aliza Rubenstein
- Institute of Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Computational Biology and Molecular Biophysics Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Marion F Sauer
- Chemical and Physical Biology Program, Vanderbilt Vaccine Center, Vanderbilt University, Nashville, TN, USA
| | - Andreas Scheck
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - William Schief
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yuval Sedan
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alexander M Sevy
- Chemical and Physical Biology Program, Vanderbilt Vaccine Center, Vanderbilt University, Nashville, TN, USA
| | - Nikolaos G Sgourakis
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Lei Shi
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Justin B Siegel
- Department of Chemistry, University of California, Davis, Davis, CA, USA
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, California, USA
- Genome Center, University of California, Davis, Davis, CA, USA
| | | | - Shannon Smith
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Yifan Song
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Cyrus Biotechnology, Seattle, WA, USA
| | - Amelie Stein
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Maria Szegedy
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Frank D Teets
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Summer B Thyme
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Ray Yu-Ruei Wang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Andrew Watkins
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Lior Zimmerman
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Richard Bonneau
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA.
- Department of Biology, New York University, New York, New York, USA.
- Department of Computer Science, New York University, New York, NY, USA.
- Center for Data Science, New York University, New York, NY, USA.
| |
Collapse
|
40
|
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: 56] [Impact Index Per Article: 11.2] [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
|
41
|
Quijano-Rubio A, Ulge UY, Walkey CD, Silva DA. The advent of de novo proteins for cancer immunotherapy. Curr Opin Chem Biol 2020; 56:119-128. [PMID: 32371023 DOI: 10.1016/j.cbpa.2020.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/22/2022]
Abstract
Engineered proteins are revolutionizing immunotherapy, but advances are still needed to harness their full potential. Traditional protein engineering methods use naturally existing proteins as a starting point, and therefore, are intrinsically limited to small alterations of a protein's natural structure and function. Conversely, computational de novo protein design is free of such limitation, and can produce a virtually infinite number of novel protein sequences, folds, and functions. Recently, we used de novo protein engineering to create Neoleukin-2/15 (Neo-2/15), a protein mimetic of the function of both interleukin-2 (IL-2) and interleukin-15 (IL-15). To our knowledge, Neo-2/15 is the first de novo protein with immunotherapeutic activity, and in murine cancer models, it has demonstrated enhanced therapeutic potency and reduced toxicity compared to IL-2. De novo protein design is already showcasing its tremendous potential for driving the next wave of protein-based therapeutics that are explicitly engineered to treat disease.
Collapse
Affiliation(s)
| | - Umut Y Ulge
- Neoleukin Therapeutics Inc., Seattle, WA, USA
| | | | | |
Collapse
|
42
|
Kuroda D, Tsumoto K. Engineering Stability, Viscosity, and Immunogenicity of Antibodies by Computational Design. J Pharm Sci 2020; 109:1631-1651. [DOI: 10.1016/j.xphs.2020.01.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/25/2019] [Accepted: 01/10/2020] [Indexed: 12/18/2022]
|
43
|
Nerli S, Sgourakis NG. Structure-based modeling of SARS-CoV-2 peptide/HLA-A02 antigens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32511353 DOI: 10.1101/2020.03.23.004176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
As a first step toward the development of diagnostic and therapeutic tools to fight the Coronavirus disease (COVID-19), it is important to characterize CD8+ T cell epitopes in the SARS-CoV-2 peptidome that can trigger adaptive immune responses. Here, we use RosettaMHC, a comparative modeling approach which leverages existing high-resolution X-ray structures from peptide/MHC complexes available in the Protein Data Bank, to derive physically realistic 3D models for high-affinity SARS-CoV-2 epitopes. We outline an application of our method to model 439 9mer and 279 10mer predicted epitopes displayed by the common allele HLA-A*02:01, and we make our models publicly available through an online database ( https://rosettamhc.chemistry.ucsc.edu ). As more detailed studies on antigen-specific T cell recognition become available, RosettaMHC models of antigens from different strains and HLA alleles can be used as a basis to understand the link between peptide/HLA complex structure and surface chemistry with immunogenicity, in the context of SARS-CoV-2 infection.
Collapse
|
44
|
Kruiswijk C, Richard G, Salverda MLM, Hindocha P, Martin WD, De Groot AS, Van Riet E. In silico identification and modification of T cell epitopes in pertussis antigens associated with tolerance. Hum Vaccin Immunother 2020; 16:277-285. [PMID: 31951773 PMCID: PMC7062413 DOI: 10.1080/21645515.2019.1703453] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The resurgence of whooping cough since the introduction of acellular (protein) vaccines has led to a renewed interest in the development of improved pertussis vaccines; Outer Membrane Vesicles (OMVs) carrying pertussis antigens have emerged as viable candidates. An in silico immunogenicity screen was carried out on 49 well-known Bordetella pertussis proteins in order to better understand their potential role toward the efficacy of pertussis OMVs for vaccine design; seven proteins were identified as being good candidates for including in optimized cellular and acellular pertussis vaccines. We then screened these antigens for putative tolerance-inducing sequences, as proteins with reduced tolerogenicity have improved vaccine potency in preclinical models. We used specialized homology tools (JanusMatrix) to identify peptides in the proteins that were cross-reactive with human sequences. Four of the 19 identified cross-reactive peptides were detolerized in silico using a separate tool, OptiMatrix, which disrupted the potential of these peptides to bind to human HLA and murine MHC. Four selected cross-reactive peptides and their detolerized variants were synthesized and their binding to a set of eight common HLA class II alleles was assessed in vitro. Reduced binding affinity to HLA class II was observed for the detolerized variants compared to the wild-type peptides, highlighting the potential of this approach for designing more efficacious pertussis vaccines.
Collapse
Affiliation(s)
- Corine Kruiswijk
- Department of Experimental Immunology & Clinical Research, Intravacc, Bilthoven, Netherlands
| | | | - Merijn L M Salverda
- Department of Experimental Immunology & Clinical Research, Intravacc, Bilthoven, Netherlands
| | | | | | | | - Elly Van Riet
- Department of Experimental Immunology & Clinical Research, Intravacc, Bilthoven, Netherlands
| |
Collapse
|
45
|
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]
|
46
|
Knudsen C, Ledsgaard L, Dehli RI, Ahmadi S, Sørensen CV, Laustsen AH. Engineering and design considerations for next-generation snakebite antivenoms. Toxicon 2019; 167:67-75. [DOI: 10.1016/j.toxicon.2019.06.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 05/22/2019] [Accepted: 06/03/2019] [Indexed: 11/27/2022]
|
47
|
Thakkar N, Bailey-Kellogg C. Balancing sensitivity and specificity in distinguishing TCR groups by CDR sequence similarity. BMC Bioinformatics 2019; 20:241. [PMID: 31092185 PMCID: PMC6521430 DOI: 10.1186/s12859-019-2864-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/29/2019] [Indexed: 12/18/2022] Open
Abstract
Background Repertoire sequencing is enabling deep explorations into the cellular immune response, including the characterization of commonalities and differences among T cell receptor (TCR) repertoires from different individuals, pathologies, and antigen specificities. In seeking to understand the generality of patterns observed in different groups of TCRs, it is necessary to balance how well each pattern represents the diversity among TCRs from one group (sensitivity) vs. how many TCRs from other groups it also represents (specificity). The variable complementarity determining regions (CDRs), particularly the third CDRs (CDR3s) interact with major histocompatibility complex (MHC)-presented epitopes from putative antigens, and thus encode the determinants of recognition. Results We here systematically characterize the predictive power that can be obtained from CDR3 sequences, using representative, readily interpretable methods for evaluating CDR sequence similarity and then clustering and classifying sequences based on similarity. An initial analysis of CDR3s of known structure, clustered by structural similarity, helps calibrate the limits of sequence diversity among CDRs that might have a common mode of interaction with presented epitopes. Subsequent analyses demonstrate that this same range of sequence similarity strikes a favorable specificity/sensitivity balance in distinguishing twins from non-twins based on overall CDR3 repertoires, classifying CDR3 repertoires by antigen specificity, and distinguishing general pathologies. Conclusion We conclude that within a fairly broad range of sequence similarity, matching CDR3 sequences are likely to share specificities. Electronic supplementary material The online version of this article (10.1186/s12859-019-2864-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Neerja Thakkar
- Department of Computer Science, Dartmouth, Hanover, NH, USA
| | | |
Collapse
|
48
|
Multifunctional CRISPR-Cas9 with engineered immunosilenced human T cell epitopes. Nat Commun 2019; 10:1842. [PMID: 31015529 PMCID: PMC6478683 DOI: 10.1038/s41467-019-09693-x] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 03/19/2019] [Indexed: 01/19/2023] Open
Abstract
The CRISPR-Cas9 system has raised hopes for developing personalized gene therapies for complex diseases. Its application for genetic and epigenetic therapies in humans raises concerns over immunogenicity of the bacterially derived Cas9 protein. Here we detect antibodies to Streptococcus pyogenes Cas9 (SpCas9) in at least 5% of 143 healthy individuals. We also report pre-existing human CD8+T cell immunity in the majority of healthy individuals screened. We identify two immunodominant SpCas9 T cell epitopes for HLA-A*02:01 using an enhanced prediction algorithm that incorporates T cell receptor contact residue hydrophobicity and HLA binding and evaluated them by T cell assays using healthy donor PBMCs. In a proof-of-principle study, we demonstrate that Cas9 protein can be modified to eliminate immunodominant epitopes through targeted mutation while preserving its function and specificity. Our study highlights the problem of pre-existing immunity against CRISPR-associated nucleases and offers a potential solution to mitigate the T cell immune response. Possible immunogenicity of the Cas9 protein raises concerns about therapeutic applications. Here the authors identify pre-existing CD8+T-cell immunity in healthy individuals and in response modify Cas9 to remove the immunodominant epitopes.
Collapse
|
49
|
Evasion of Pre-Existing Immunity to Cas9: a Prerequisite for Successful Genome Editing In Vivo? CURRENT TRANSPLANTATION REPORTS 2019. [DOI: 10.1007/s40472-019-00237-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
50
|
Diupotex M, Martínez-Salazar MB, Escalona-Montaño AR, Zamora-Chimal J, Salaiza-Suazo N, Ruiz-Remigio A, Roldán-Salgado A, Aguirre-García MM, Martínez-Calvillo S, Gaytán P, Becker I. The mKate fluorescent protein expressed by Leishmania mexicana modifies the parasite immunopathogenicity in BALB/c mice. Parasite Immunol 2019; 41:e12608. [PMID: 30500992 DOI: 10.1111/pim.12608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 11/27/2022]
Abstract
Parasites have been engineered to express fluorescent reporter proteins, yet the impact of red fluorescent proteins on Leishmania infections remains largely unknown. We analysed the infection outcome of Leishmania mexicana parasites engineered for the constitutive expression of mKate protein and evaluated their immunogenicity in BALB/c mice. Infection of BALB/c mice with mKate transfected L. mexicana (LmexmKate ) parasites caused enlarged lesion sizes, leading to ulceration, and containing more parasites, as compared to LmexWT . The mKate protein showed immunogenic properties inducing antibody production against the mKate protein, as well as enhancing antibody production against the parasite. The augmented lesion sizes and ulcers, together with the more elevated antibody production, were related to an enhanced number of TNF-α and IL-1β producing cells in the infected tissues. We conclude that mKate red fluorescent protein is an immunogenic protein, capable of modifying disease evolution of L. mexicana.
Collapse
Affiliation(s)
- Mariana Diupotex
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Ciudad de México, México
| | - María Berenice Martínez-Salazar
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Ciudad de México, México
| | - Alma Reyna Escalona-Montaño
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Ciudad de México, México
| | - Jaime Zamora-Chimal
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Ciudad de México, México
| | - Norma Salaiza-Suazo
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Ciudad de México, México
| | - Adriana Ruiz-Remigio
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Ciudad de México, México
| | | | - María Magdalena Aguirre-García
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Ciudad de México, México
| | - Santiago Martínez-Calvillo
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, México
| | - Paul Gaytán
- Instituto de Biotecnología-Universidad Nacional Autónoma de México, Cuernavaca, México
| | - Ingeborg Becker
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Ciudad de México, México
| |
Collapse
|