1
|
Dobersberger M, Sumesgutner D, Zajc CU, Traxlmayr MW. Protocol for engineering binding domains to recognize ligand-bound receptors by using yeast surface display. STAR Protoc 2024; 5:103339. [PMID: 39321026 PMCID: PMC11462054 DOI: 10.1016/j.xpro.2024.103339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/01/2024] [Accepted: 09/04/2024] [Indexed: 09/27/2024] Open
Abstract
Yeast surface display is a versatile protein engineering technology, enabling precise control of the applied selection pressure. We present a yeast-surface-display-based protocol for the enrichment of binders specifically recognizing ligand-bound receptors. We describe steps for magnetic bead selections, random mutagenesis, and flow cytometric sorting, followed by library sequencing and detailed analysis of enriched clones. While this approach is exemplified with rcSso7d-based libraries and epidermal growth factor (EGF)-epidermal growth factor receptor (EGFR) complexes, it can also be adapted to other binder scaffolds and ligand-receptor systems. For complete details on the use and execution of this protocol, please refer to Dobersberger et al.1.
Collapse
Affiliation(s)
- Markus Dobersberger
- Department of Chemistry, Institute of Biochemistry, BOKU University, Vienna 1190, Austria
| | - Delia Sumesgutner
- Department of Chemistry, Institute of Biochemistry, BOKU University, Vienna 1190, Austria; CD Laboratory for Next Generation CAR T Cells, Vienna 1090, Austria.
| | - Charlotte U Zajc
- Department of Chemistry, Institute of Biochemistry, BOKU University, Vienna 1190, Austria; CD Laboratory for Next Generation CAR T Cells, Vienna 1090, Austria
| | - Michael W Traxlmayr
- Department of Chemistry, Institute of Biochemistry, BOKU University, Vienna 1190, Austria; CD Laboratory for Next Generation CAR T Cells, Vienna 1090, Austria.
| |
Collapse
|
2
|
Chen M, Corless EI, Engelward BP, Swager TM. Optical Detection of Interleukin-6 Using Liquid Janus Emulsions Using Hyperthermophilic Affinity Proteins. ACS OMEGA 2024; 9:37076-37085. [PMID: 39246480 PMCID: PMC11375700 DOI: 10.1021/acsomega.4c03959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 09/10/2024]
Abstract
When equal volumes of two immiscible liquids are mixed (e.g., a hydrocarbon and a fluorocarbon), Janus droplets can form in an aqueous solution. In a gravity-aligned Janus droplet, the boundary between the two phases is flat and thus optically transparent when viewed from above. When tipped due to interactions with an analyte (i.e., agglutination), the resulting change in refraction and reflection yields an optical signal that can be detected and quantified. This study reports the detection and quantitation of interleukin-6 (IL-6) using emulsions functionalized at the hydrocarbon:aqueous interface with engineered proteins that bind IL-6 at high affinity and specificity. Hyperthermophilic affinity proteins (rcSso7d) are derived from thermophiles, giving them excellent thermal stability. Two rcSso7d affinity protein variants were synthesized with a noncanonical azide-functionalized amino acid to enable click chemistry to novel polymeric anchors embedded in the hydrocarbon phase. The two binding proteins recognize different epitopes, enabling the detection of both monomeric and dimeric IL-6 via agglutination. It is noteworthy that the rsSso7d protein variants, in addition to having superior thermal stability and facile recombinant synthesis in E. coli, show superior performance when compared to commercial antibodies for IL-6.
Collapse
Affiliation(s)
- Michelle Chen
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| | - Elliot I. Corless
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Bevin P. Engelward
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Timothy M. Swager
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
3
|
Dobersberger M, Sumesgutner D, Zajc CU, Salzer B, Laurent E, Emminger D, Sylvander E, Lehner E, Teufl M, Seigner J, Bobbili MR, Kunert R, Lehner M, Traxlmayr MW. An engineering strategy to target activated EGFR with CAR T cells. CELL REPORTS METHODS 2024; 4:100728. [PMID: 38492569 PMCID: PMC11045874 DOI: 10.1016/j.crmeth.2024.100728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/18/2024] [Accepted: 02/16/2024] [Indexed: 03/18/2024]
Abstract
Chimeric antigen receptor (CAR) T cells have shown remarkable response rates in hematological malignancies. In contrast, CAR T cell treatment of solid tumors is associated with several challenges, in particular the expression of most tumor-associated antigens at lower levels in vital organs, resulting in on-target/off-tumor toxicities. Thus, innovative approaches to improve the tumor specificity of CAR T cells are urgently needed. Based on the observation that many human solid tumors activate epidermal growth factor receptor (EGFR) on their surface through secretion of EGFR ligands, we developed an engineering strategy for CAR-binding domains specifically directed against the ligand-activated conformation of EGFR. We show, in several experimental systems, that the generated binding domains indeed enable CAR T cells to distinguish between active and inactive EGFR. We anticipate that this engineering concept will be an important step forward to improve the tumor specificity of CAR T cells directed against EGFR-positive solid cancers.
Collapse
Affiliation(s)
- Markus Dobersberger
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria
| | - Delia Sumesgutner
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria; CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria
| | - Charlotte U Zajc
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria; CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria
| | - Benjamin Salzer
- CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria; St. Anna Children's Cancer Research Institute, CCRI, 1090 Vienna, Austria
| | - Elisabeth Laurent
- BOKU Core Facility Biomolecular & Cellular Analysis, BOKU University, 1190 Vienna, Austria
| | - Dominik Emminger
- CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria; St. Anna Children's Cancer Research Institute, CCRI, 1090 Vienna, Austria
| | - Elise Sylvander
- CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria; St. Anna Children's Cancer Research Institute, CCRI, 1090 Vienna, Austria
| | - Elisabeth Lehner
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria; CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria
| | - Magdalena Teufl
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria; CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria
| | - Jacqueline Seigner
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria; Department of Biotechnology, Institute of Animal Cell Technology and Systems Biology, BOKU University, 1190 Vienna, Austria
| | - Madhusudhan Reddy Bobbili
- Department of Biotechnology, Institute of Molecular Biotechnology, BOKU University, 1190 Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, Research Center in Cooperation with AUVA, 1200 Vienna, Austria
| | - Renate Kunert
- Department of Biotechnology, Institute of Animal Cell Technology and Systems Biology, BOKU University, 1190 Vienna, Austria
| | - Manfred Lehner
- CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria; St. Anna Children's Cancer Research Institute, CCRI, 1090 Vienna, Austria; St. Anna Children's Hospital, Department of Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael W Traxlmayr
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria; CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria.
| |
Collapse
|
4
|
Meksiriporn B, Spangler JB. Directed-evolution approach to empower EGFR targeting for immunotherapy. CELL REPORTS METHODS 2024; 4:100762. [PMID: 38631347 PMCID: PMC11046029 DOI: 10.1016/j.crmeth.2024.100762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/01/2024] [Accepted: 04/01/2024] [Indexed: 04/19/2024]
Abstract
Advances in directed-evolution technologies are enabling new strategies to isolate binding proteins that recognize disease-associated states of a target protein. In this issue of Cell Reports Methods, Dobersberger et al. devised a yeast display-based selection scheme to discover proteins that engage the cancer-associated activated state of a receptor to enable design of safe and effective immunotherapies.
Collapse
Affiliation(s)
- Bunyarit Meksiriporn
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biology, School of Science, King Mongkut's Institute of Technology Ladkrabang, Bangkok 10520, Thailand
| | - Jamie B Spangler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Molecular Microbiology & Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
5
|
Xu X, Xie M, Luo S, Jia X. Revisiting Protein-Copolymer Binding Mechanisms: Insights beyond the "Lock-and-Key" Model. J Phys Chem Lett 2024; 15:773-781. [PMID: 38227953 DOI: 10.1021/acs.jpclett.3c03200] [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: 01/18/2024]
Abstract
The "lock-and-key" model that emphasizes the concept of chemical-structural complementary is the key mechanism for explaining the selectivity between small ligands and a larger adsorbent molecule. In this work, concerning the copolymer chain using only the combination of N-isopropylacrylamide (NIPAm) and hydrophobic N-tert-butylacrylamide (TBAm) monomers and by large-scale atomistic molecular dynamics simulations, our results show that the flexible copolymer chain may exhibit strong binding affinity for the biomarker protein epithelial cell adhesion molecule, in the absence of hydrophobic matching and strong structural complementarity. This surprising binding behavior, which cannot be anticipated by the "lock-and-key" model, can be attributed to the preferential interactions established by the copolymer with the protein's hydrophilic exterior. We observe that increasing the fraction of incorporated TBAm monomers leads to a prevalence of interactions with asparagine and glutamine amino acids due to the emerging hydrogen bonding with both NIPAm and TBAm monomers. Our findings suggest the appearance of highly specific and high-affinity binding sites on the protein created by engineering the copolymer composition, which motivates the applications of copolymers as protein affinity reagents.
Collapse
Affiliation(s)
- Xiao Xu
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei, Nanjing 210094, P. R. China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Menghan Xie
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei, Nanjing 210094, P. R. China
| | - Shejia Luo
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei, Nanjing 210094, P. R. China
| | - Xu Jia
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei, Nanjing 210094, P. R. China
| |
Collapse
|
6
|
Momin N. Balancing safety and efficacy: tuning the biodistribution and pharmacokinetics of cytokine immunotherapies. Curr Opin Biotechnol 2023; 84:102994. [PMID: 37806081 DOI: 10.1016/j.copbio.2023.102994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/12/2023] [Accepted: 08/27/2023] [Indexed: 10/10/2023]
Abstract
Modulating the immune system shows promise in treating various conditions such as autoimmune, malignant, inflammatory, and infectious diseases. While immunotherapies can provide significant clinical benefits, they can also trigger debilitating immune-related toxicities. Achieving a balance between safety and efficacy of immunotherapy remains a significant engineering challenge. A complex immune response can be simplified into a sequence of coordinated signals with precise spatial and temporal arrangements. Mimicking or inhibiting these signals with protein immunotherapies relies on engineering them with specific biodistribution and pharmacokinetic properties. This review summarizes principles governing the movement of therapeutic proteins (i.e. biologics), focusing on cytokine immunotherapies injected intravenously or locally, and highlights approaches and considerations to balance their efficacy and safety.
Collapse
Affiliation(s)
- Noor Momin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Center for Precision Engineering for Health, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Lopez-Martinez E, Manteca A, Ferruz N, Cortajarena AL. Statistical Analysis and Tokenization of Epitopes to Construct Artificial Neoepitope Libraries. ACS Synth Biol 2023; 12:2812-2818. [PMID: 37703075 PMCID: PMC10594869 DOI: 10.1021/acssynbio.3c00201] [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: 03/31/2023] [Indexed: 09/14/2023]
Abstract
Epitopes are specific regions on an antigen's surface that the immune system recognizes. Epitopes are usually protein regions on foreign immune-stimulating entities such as viruses and bacteria, and in some cases, endogenous proteins may act as antigens. Identifying epitopes is crucial for accelerating the development of vaccines and immunotherapies. However, mapping epitopes in pathogen proteomes is challenging using conventional methods. Screening artificial neoepitope libraries against antibodies can overcome this issue. Here, we applied conventional sequence analysis and methods inspired in natural language processing to reveal specific sequence patterns in the linear epitopes deposited in the Immune Epitope Database (www.iedb.org) that can serve as building blocks for the design of universal epitope libraries. Our results reveal that amino acid frequency in annotated linear epitopes differs from that in the human proteome. Aromatic residues are overrepresented, while the presence of cysteines is practically null in epitopes. Byte pair encoding tokenization shows high frequencies of tryptophan in tokens of 5, 6, and 7 amino acids, corroborating the findings of the conventional sequence analysis. These results can be applied to reduce the diversity of linear epitope libraries by orders of magnitude.
Collapse
Affiliation(s)
- Elena Lopez-Martinez
- Centre
for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014 Spain
| | - Aitor Manteca
- Centre
for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014 Spain
| | - Noelia Ferruz
- Molecular
Biology Institute of Barcelona (IBMB-CSIC), Barcelona Science Park, Baldiri Reixac, 15-21, 08028, Barcelona, Spain
| | - Aitziber L. Cortajarena
- Centre
for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014 Spain
- IKERBASQUE, Basque
Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain
| |
Collapse
|
8
|
Morris MA, Mills CE, Paloni JM, Miller EA, Sikes HD, Olsen BD. High-Throughput Screening of Streptavidin-Binding Proteins in Self-Assembled Solid Films for Directed Evolution of Materials. NANO LETTERS 2023; 23:7303-7310. [PMID: 37566825 DOI: 10.1021/acs.nanolett.3c01229] [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: 08/13/2023]
Abstract
Evolution has shaped the development of proteins with an incredible diversity of properties. Incorporating proteins into materials is desirable for applications including biosensing; however, high-throughput selection techniques for screening protein libraries in materials contexts is lacking. In this work, a high-throughput platform to assess the binding affinity for ordered sensing proteins was established. A library of fusion proteins, consisting of an elastin-like polypeptide block, one of 22 variants of rcSso7d, and a coiled-coil order-directing sequence, was generated. All selected variants had high binding in films, likely due to the similarity of the assay to magnetic bead sorting used for initial selection, while solution binding was more variable. From these results, both the assembly of the fusion proteins in their operating state and the functionality of the binding protein are key factors in the biosensing performance. Thus, the integration of directed evolution with assembled systems is necessary to the design of better materials.
Collapse
Affiliation(s)
- Melody A Morris
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Carolyn E Mills
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Justin M Paloni
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Eric A Miller
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Hadley D Sikes
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Bradley D Olsen
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
9
|
Lax BM, Palmeri JR, Lutz EA, Sheen A, Stinson JA, Duhamel L, Santollani L, Kennedy A, Rothschilds AM, Spranger S, Sansom DM, Wittrup KD. Both intratumoral regulatory T cell depletion and CTLA-4 antagonism are required for maximum efficacy of anti-CTLA-4 antibodies. Proc Natl Acad Sci U S A 2023; 120:e2300895120. [PMID: 37487077 PMCID: PMC10400942 DOI: 10.1073/pnas.2300895120] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 06/23/2023] [Indexed: 07/26/2023] Open
Abstract
Anti-CTLA-4 antibodies have successfully elicited durable tumor regression in the clinic; however, long-term benefit is limited to a subset of patients for select cancer indications. The incomplete understanding of their mechanism of action has hindered efforts at improvement, with conflicting hypotheses proposing either antagonism of the CTLA-4:B7 axis or Fc effector-mediated regulatory T cell (Treg) depletion governing efficacy. Here, we report the engineering of a nonantagonistic CTLA-4 binding domain (b1s1e2) that depletes intratumoral Tregs as an Fc fusion. Comparison of b1s1e2-Fc to 9d9, an antagonistic anti-CTLA-4 antibody, allowed for interrogation of the separate contributions of CTLA-4 antagonism and Treg depletion to efficacy. Despite equivalent levels of intratumoral Treg depletion, 9d9 achieved more long-term cures than b1s1e2-Fc in MC38 tumors, demonstrating that CTLA-4 antagonism provided additional survival benefit. Consistent with prior reports that CTLA-4 antagonism enhances priming, treatment with 9d9, but not b1s1e2-Fc, increased the percentage of activated T cells in the tumor-draining lymph node (tdLN). Treg depletion with either construct was restricted to the tumor due to insufficient surface CTLA-4 expression on Tregs in other compartments. Through intratumoral administration of diphtheria toxin in Foxp3-DTR mice, we show that depletion of both intratumoral and nodal Tregs provided even greater survival benefit than 9d9, consistent with Treg-driven restraint of priming in the tdLN. Our data demonstrate that anti-CTLA-4 therapies require both CTLA-4 antagonism and intratumoral Treg depletion for maximum efficacy-but that potential future therapies also capable of depleting nodal Tregs could show efficacy in the absence of CTLA-4 antagonism.
Collapse
Affiliation(s)
- Brianna M. Lax
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Joseph R. Palmeri
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Emi A. Lutz
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Allison Sheen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Jordan A. Stinson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Lauren Duhamel
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Luciano Santollani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Alan Kennedy
- Institute of Immunity and Transplantation, University College London, LondonNW3 2PP, United Kingdom
| | - Adrienne M. Rothschilds
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Stefani Spranger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - David M. Sansom
- Institute of Immunity and Transplantation, University College London, LondonNW3 2PP, United Kingdom
| | - K. Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| |
Collapse
|
10
|
Ward EM, Zamora CY, Schocker NS, Ghosh S, Kizer ME, Imperiali B. Engineered Glycan-Binding Proteins for Recognition of the Thomsen-Friedenreich Antigen and Structurally Related Disaccharides. ACS Chem Biol 2023; 18:70-80. [PMID: 36525666 PMCID: PMC9868099 DOI: 10.1021/acschembio.2c00683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Glycan-binding proteins (GBPs) are widely used reagents for basic research and clinical applications. These reagents allow for the identification and manipulation of glycan determinants without specialized equipment or time-consuming experimental methods. Existing GBPs, mainly antibodies and lectins, are limited, and discovery or creation of reagents with novel specificities is time consuming and difficult. Here, we detail the generation of GBPs from a small, hyper-thermostable DNA-binding protein by directed evolution. Yeast surface display of a variable library of rcSso7d proteins was screened to find variants with selectivity toward the cancer-associated glycan Galβ1-3GalNAcα or Thomsen-Friedenreich antigen and various relevant disaccharides. Characterization of these proteins shows them to have specificities and affinities on par with currently available lectins. The proteins can be readily functionalized with fluorophores or biotin using sortase-mediated ligation to create reagents that prove useful for glycoprotein blotting and cell staining applications. The presented methods for the development of GBPs toward specific saccharides of interest will have great impact on both biomedical and glycobiological research.
Collapse
Affiliation(s)
- Elizabeth M. Ward
- Department of Biology, Massachusetts Institute of Technology, 31 Ames St, Cambridge, MA 02142, USA,Microbiology Graduate Program, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Cristina Y. Zamora
- Department of Biology, Massachusetts Institute of Technology, 31 Ames St, Cambridge, MA 02142, USA,Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Nathaniel S. Schocker
- Department of Biology, Massachusetts Institute of Technology, 31 Ames St, Cambridge, MA 02142, USA,Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Soumi Ghosh
- Department of Biology, Massachusetts Institute of Technology, 31 Ames St, Cambridge, MA 02142, USA,Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Megan E. Kizer
- Department of Biology, Massachusetts Institute of Technology, 31 Ames St, Cambridge, MA 02142, USA,Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Barbara Imperiali
- Department of Biology, Massachusetts Institute of Technology, 31 Ames St, Cambridge, MA 02142, USA,Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA,Corresponding author
| |
Collapse
|
11
|
Teufl M, Zajc CU, Traxlmayr MW. Engineering Strategies to Overcome the Stability-Function Trade-Off in Proteins. ACS Synth Biol 2022; 11:1030-1039. [PMID: 35258287 PMCID: PMC8938945 DOI: 10.1021/acssynbio.1c00512] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
In addition to its
biological function, the stability of a protein
is a major determinant for its applicability. Unfortunately, engineering
proteins for improved functionality usually results in destabilization
of the protein. This so-called stability–function trade-off
can be explained by the simple fact that the generation of a novel
protein function—or the improvement of an existing one—necessitates
the insertion of mutations, i.e., deviations from
the evolutionarily optimized wild-type sequence. In fact, it was demonstrated
that gain-of-function mutations are not more destabilizing than other
random mutations. The stability–function trade-off is a universal
phenomenon during protein evolution that has been observed with completely
different types of proteins, including enzymes, antibodies, and engineered
binding scaffolds. In this review, we discuss three types of strategies
that have been successfully deployed to overcome this omnipresent
obstacle in protein engineering approaches: (i) using highly stable
parental proteins, (ii) minimizing the extent of destabilization during
functional engineering (by library optimization and/or coselection
for stability and function), and (iii) repairing damaged mutants through
stability engineering. The implementation of these strategies in protein
engineering campaigns will facilitate the efficient generation of
protein variants that are not only functional but also stable and
therefore better-suited for subsequent applications.
Collapse
Affiliation(s)
- Magdalena Teufl
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, 1190 Vienna, Austria
- CD Laboratory for Next Generation CAR T Cells, 1190 Vienna, Austria
| | - Charlotte U. Zajc
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, 1190 Vienna, Austria
- CD Laboratory for Next Generation CAR T Cells, 1190 Vienna, Austria
| | - Michael W. Traxlmayr
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, 1190 Vienna, Austria
- CD Laboratory for Next Generation CAR T Cells, 1190 Vienna, Austria
| |
Collapse
|
12
|
Kang BH, Lax BM, Wittrup KD. Yeast Surface Display for Protein Engineering: Library Generation, Screening, and Affinity Maturation. Methods Mol Biol 2022; 2491:29-62. [PMID: 35482183 DOI: 10.1007/978-1-0716-2285-8_2] [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: 06/14/2023]
Abstract
Yeast surface display is a powerful directed evolution method for developing and engineering protein molecules to attain desired properties. Here, updated protocols are presented for purposes of identification of lead binders and their affinity maturation. Large libraries are screened by magnetic bead selections followed by flow cytometric selections. Upon identification and characterization of single clones, their affinities are improved by an iterative process of mutagenesis and fluorescence-activated cell sorting.
Collapse
Affiliation(s)
- Byong H Kang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brianna M Lax
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - K Dane Wittrup
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
13
|
Corless E, Hao Y, Jia H, Kongsuphol P, Tay DMY, Ng SY, Sikes HD. Generation of Thermally Stable Affinity Pairs for Sensitive, Specific Immunoassays. Methods Mol Biol 2022; 2491:417-469. [PMID: 35482202 DOI: 10.1007/978-1-0716-2285-8_21] [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] [Indexed: 06/14/2023]
Abstract
Many point-of-care diagnostic tests rely on a pair of monoclonal antibodies that bind to two distinct epitopes of a molecule of interest. This protocol describes the identification and generation of such affinity pairs based on an easily produced small protein scaffold rcSso7d which can substitute monoclonal antibodies. These strong binding variants are identified from a large yeast display library. The approach described can be significantly faster than antibody generation and epitope binning, yielding affinity pairs synthesized in common bacterial protein synthesis strains, enabling the rapid generation of novel diagnostic tools.
Collapse
Affiliation(s)
- Elliot Corless
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yining Hao
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Huan Jia
- Antimicrobial Resistance Interdisciplinary Research Group (AMR-IRG), Singapore-MIT Alliance in Research and Technology (SMART), Singapore, Singapore
| | - Patthara Kongsuphol
- Antimicrobial Resistance Interdisciplinary Research Group (AMR-IRG), Singapore-MIT Alliance in Research and Technology (SMART), Singapore, Singapore
| | - Dousabel M Y Tay
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Say Yong Ng
- Antimicrobial Resistance Interdisciplinary Research Group (AMR-IRG), Singapore-MIT Alliance in Research and Technology (SMART), Singapore, Singapore
| | - Hadley D Sikes
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Antimicrobial Resistance Interdisciplinary Research Group (AMR-IRG), Singapore-MIT Alliance in Research and Technology (SMART), Singapore, Singapore.
| |
Collapse
|
14
|
Abstract
Yeast surface display is a powerful protein engineering technology that is extensively used to improve various properties of proteins, including affinity, specificity, and stability or even to add novel functions (usually ligand binding). Apart from its robustness and versatility as an engineering tool, yeast display offers a further critical advantage: Once the selection campaign is finished, usually resulting in an oligoclonal pool, these enriched protein variants can be analyzed individually on the surface of yeast without the need for any sub-cloning, soluble expression, and purification. Here, we provide detailed protocols for determining both the affinity and the thermal stability of yeast displayed proteins. In addition, we discuss the advantages, challenges, and potential pitfalls associated with affinity and stability analysis using yeast surface display.
Collapse
Affiliation(s)
- Charlotte U Zajc
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, Vienna, Austria
- CD Laboratory for Next Generation CAR T Cells, Vienna, Austria
| | - Magdalena Teufl
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, Vienna, Austria
- CD Laboratory for Next Generation CAR T Cells, Vienna, Austria
| | - Michael W Traxlmayr
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, Vienna, Austria.
| |
Collapse
|
15
|
Paumann-Page M, Kienzl NF, Motwani J, Bathish B, Paton LN, Magon NJ, Sevcnikar B, Furtmüller PG, Traxlmayr MW, Obinger C, Eccles MR, Winterbourn CC. Peroxidasin protein expression and enzymatic activity in metastatic melanoma cell lines are associated with invasive potential. Redox Biol 2021; 46:102090. [PMID: 34438259 PMCID: PMC8390535 DOI: 10.1016/j.redox.2021.102090] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 07/31/2021] [Indexed: 02/06/2023] Open
Abstract
Peroxidasin, a heme peroxidase, has been shown to play a role in cancer progression. mRNA expression has been reported to be upregulated in metastatic melanoma cell lines and connected to the invasive phenotype, but little is known about how peroxidasin acts in cancer cells. We have analyzed peroxidasin protein expression and activity in eight metastatic melanoma cell lines using an ELISA developed with an in-house peroxidasin binding protein. RNAseq data analysis confirmed high peroxidasin mRNA expression in the five cell lines classified as invasive and low expression in the three non-invasive cell lines. Protein levels of peroxidasin were higher in the cell lines with an invasive phenotype. Active peroxidasin was secreted to the cell culture medium, where it accumulated over time, and peroxidasin protein levels in the medium were also much higher in invasive than non-invasive cell lines. The only well-established physiological role of peroxidasin is in the formation of a sulfilimine bond, which cross-links collagen IV in basement membranes via catalyzed oxidation of bromide to hypobromous acid. We found that peroxidasin secreted from melanoma cells formed sulfilimine bonds in uncross-linked collagen IV, confirming peroxidasin activity and hypobromous acid formation. Moreover, 3-bromotyrosine, a stable product of hypobromous acid reacting with tyrosine residues, was detected in invasive melanoma cells, substantiating that their expression of peroxidasin generates hypobromous acid, and showing that it does not exclusively react with collagen IV, but also with other biomolecules.
Collapse
Affiliation(s)
- Martina Paumann-Page
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago Christchurch, P.O. Box 4345, Christchurch, New Zealand.
| | - Nikolaus F Kienzl
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
| | - Jyoti Motwani
- Department of Pathology, Dunedin School of Medicine, University of Otago, 270 Great King Street, Dunedin, 9054, New Zealand
| | - Boushra Bathish
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago Christchurch, P.O. Box 4345, Christchurch, New Zealand
| | - Louise N Paton
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago Christchurch, P.O. Box 4345, Christchurch, New Zealand
| | - Nicholas J Magon
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago Christchurch, P.O. Box 4345, Christchurch, New Zealand
| | - Benjamin Sevcnikar
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
| | - Paul G Furtmüller
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
| | - Michael W Traxlmayr
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
| | - Christian Obinger
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
| | - Mike R Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, 270 Great King Street, Dunedin, 9054, New Zealand
| | - Christine C Winterbourn
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago Christchurch, P.O. Box 4345, Christchurch, New Zealand
| |
Collapse
|
16
|
Abstract
RAS proteins represent critical drivers of tumor development and thus are the focus of intense efforts to pharmacologically inhibit these proteins in human cancer. Although recent success has been attained in developing clinically efficacious inhibitors to KRASG12C, there remains a critical need for developing approaches to inhibit additional mutant RAS proteins. A number of anti-RAS biologics have been developed which reveal novel and potentially therapeutically targetable vulnerabilities in oncogenic RAS. This review will discuss the growing field of anti-RAS biologics and potential development of these reagents into new anti-RAS therapies.
Collapse
Affiliation(s)
- Michael Whaby
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Imran Khan
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States
| | - John P O'Bryan
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States.
| |
Collapse
|
17
|
Laurent E, Sieber A, Salzer B, Wachernig A, Seigner J, Lehner M, Geyeregger R, Kratzer B, Jäger U, Kunert R, Pickl WF, Traxlmayr MW. Directed Evolution of Stabilized Monomeric CD19 for Monovalent CAR Interaction Studies and Monitoring of CAR-T Cell Patients. ACS Synth Biol 2021; 10:1184-1198. [PMID: 33843201 PMCID: PMC8155657 DOI: 10.1021/acssynbio.1c00010] [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] [Indexed: 12/19/2022]
Abstract
CD19 is among the most relevant targets in cancer immunotherapy. However, its extracellular domain (ECD) is prone to aggregation and misfolding, representing a major obstacle for the development and analysis of CD19-targeted therapeutics. Here, we engineered stabilized CD19-ECD (termed SuperFolder) variants, which also showed improved expression rates and, in contrast to the wild type protein, they could be efficiently purified in their monomeric forms. Despite being considerably more stable, these engineered mutants largely preserved the wild type sequence (>98.8%). We demonstrate that the variant SF05 enabled the determination of the monovalent affinity between CD19 and a clinically approved FMC63-based CAR, as well as monitoring and phenotypic characterization of CD19-directed CAR-T cells in the blood of lymphoma patients. We anticipate that the SuperFolder mutants generated in this study will be highly valuable tools for a range of applications in basic immunology and CD19-targeted cancer immunotherapy.
Collapse
Affiliation(s)
- Elisabeth Laurent
- Department of Biotechnology and BOKU Core Facility Biomolecular and Cellular Analysis, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Anna Sieber
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Benjamin Salzer
- St. Anna Children’s Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - Anna Wachernig
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Jacqueline Seigner
- Department of Chemistry, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Manfred Lehner
- St. Anna Children’s Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - René Geyeregger
- St. Anna Children’s Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, 1090 Vienna, Austria
| | - Ulrich Jäger
- Department of Internal Medicine, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Renate Kunert
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Winfried F. Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, 1090 Vienna, Austria
| | - Michael W. Traxlmayr
- Department of Chemistry, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| |
Collapse
|
18
|
Tilegenova C, Izadi S, Yin J, Huang CS, Wu J, Ellerman D, Hymowitz SG, Walters B, Salisbury C, Carter PJ. Dissecting the molecular basis of high viscosity of monospecific and bispecific IgG antibodies. MAbs 2021; 12:1692764. [PMID: 31779513 PMCID: PMC6927759 DOI: 10.1080/19420862.2019.1692764] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Some antibodies exhibit elevated viscosity at high concentrations, making them poorly suited for therapeutic applications requiring administration by injection such as subcutaneous or ocular delivery. Here we studied an anti-IL-13/IL-17 bispecific IgG4 antibody, which has anomalously high viscosity compared to its parent monospecific antibodies. The viscosity of the bispecific IgG4 in solution was decreased by only ~30% in the presence of NaCl, suggesting electrostatic interactions are insufficient to fully explain the drivers of viscosity. Intriguingly, addition of arginine-HCl reduced the viscosity of the bispecific IgG4 by ~50% to its parent IgG level. These data suggest that beyond electrostatics, additional types of interactions such as cation-π and/or π-π may contribute to high viscosity more significantly than previously understood. Molecular dynamics simulations of antibody fragments in the mixed solution of free arginine and explicit water were conducted to identify hotspots involved in self-interactions. Exposed surface aromatic amino acids displayed an increased number of contacts with arginine. Mutagenesis of the majority of aromatic residues pinpointed by molecular dynamics simulations effectively decreased the solution's viscosity when tested experimentally. This mutational method to reduce the viscosity of a bispecific antibody was extended to a monospecific anti-GCGR IgG1 antibody with elevated viscosity. In all cases, point mutants were readily identified that both reduced viscosity and retained antigen-binding affinity. These studies demonstrate a new approach to mitigate high viscosity of some antibodies by mutagenesis of surface-exposed aromatic residues on complementarity-determining regions that may facilitate some clinical applications.
Collapse
Affiliation(s)
| | - Saeed Izadi
- Early Stage Pharmaceutical Development, Genentech Inc., South San Francisco, CA, USA
| | - Jianping Yin
- Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | | | - Jiansheng Wu
- Protein Chemistry, Genentech Inc., South San Francisco, CA, USA
| | - Diego Ellerman
- Protein Chemistry, Genentech Inc., South San Francisco, CA, USA
| | - Sarah G Hymowitz
- Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | - Benjamin Walters
- Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Cleo Salisbury
- Early Stage Pharmaceutical Development, Genentech Inc., South San Francisco, CA, USA
| | - Paul J Carter
- Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| |
Collapse
|
19
|
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: 18] [Impact Index Per Article: 4.5] [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
|
20
|
Salzer B, Schueller CM, Zajc CU, Peters T, Schoeber MA, Kovacic B, Buri MC, Lobner E, Dushek O, Huppa JB, Obinger C, Putz EM, Holter W, Traxlmayr MW, Lehner M. Engineering AvidCARs for combinatorial antigen recognition and reversible control of CAR function. Nat Commun 2020; 11:4166. [PMID: 32820173 PMCID: PMC7441178 DOI: 10.1038/s41467-020-17970-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 07/23/2020] [Indexed: 12/13/2022] Open
Abstract
T cells engineered to express chimeric antigen receptors (CAR-T cells) have shown impressive clinical efficacy in the treatment of B cell malignancies. However, the development of CAR-T cell therapies for solid tumors is hampered by the lack of truly tumor-specific antigens and poor control over T cell activity. Here we present an avidity-controlled CAR (AvidCAR) platform with inducible and logic control functions. The key is the combination of (i) an improved CAR design which enables controlled CAR dimerization and (ii) a significant reduction of antigen-binding affinities to introduce dependence on bivalent interaction, i.e. avidity. The potential and versatility of the AvidCAR platform is exemplified by designing ON-switch CARs, which can be regulated with a clinically applied drug, and AND-gate CARs specifically recognizing combinations of two antigens. Thus, we expect that AvidCARs will be a highly valuable platform for the development of controllable CAR therapies with improved tumor specificity.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Cells, Cultured
- Cytokines/immunology
- Cytokines/metabolism
- Cytotoxicity, Immunologic/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Lymphocyte Activation/immunology
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Benjamin Salzer
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
- Christian Doppler Laboratory for Next Generation CAR T Cells, 1090, Vienna, Austria
| | | | - Charlotte U Zajc
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
- Christian Doppler Laboratory for Next Generation CAR T Cells, 1090, Vienna, Austria
| | - Timo Peters
- Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael A Schoeber
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
| | - Boris Kovacic
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
| | - Michelle C Buri
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
| | - Elisabeth Lobner
- Department of Biotechnology, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Johannes B Huppa
- Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Christian Obinger
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
| | - Eva M Putz
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
| | - Wolfgang Holter
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
- Department of Pediatrics, St. Anna Kinderspital, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael W Traxlmayr
- Christian Doppler Laboratory for Next Generation CAR T Cells, 1090, Vienna, Austria.
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria.
| | - Manfred Lehner
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria.
- Christian Doppler Laboratory for Next Generation CAR T Cells, 1090, Vienna, Austria.
- Department of Pediatrics, St. Anna Kinderspital, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
21
|
A conformation-specific ON-switch for controlling CAR T cells with an orally available drug. Proc Natl Acad Sci U S A 2020; 117:14926-14935. [PMID: 32554495 PMCID: PMC7334647 DOI: 10.1073/pnas.1911154117] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Molecular ON-switches in which a chemical compound induces protein-protein interactions can allow cellular function to be controlled with small molecules. ON-switches based on clinically applicable compounds and human proteins would greatly facilitate their therapeutic use. Here, we developed an ON-switch system in which the human retinol binding protein 4 (hRBP4) of the lipocalin family interacts with engineered hRBP4 binders in a small molecule-dependent manner. Two different protein scaffolds were engineered to bind to hRBP4 when loaded with the orally available small molecule A1120. The crystal structure of an assembled ON-switch shows that the engineered binder specifically recognizes the conformational changes induced by A1120 in two loop regions of hRBP4. We demonstrate that this conformation-specific ON-switch is highly dependent on the presence of A1120, as demonstrated by an ∼500-fold increase in affinity upon addition of the small molecule drug. Furthermore, the ON-switch successfully regulated the activity of primary human CAR T cells in vitro. We anticipate that lipocalin-based ON-switches have the potential to be broadly applied for the safe pharmacological control of cellular therapeutics.
Collapse
|
22
|
Sung KJ, Jabbour Al Maalouf Y, Johns QR, Miller EA, Sikes HD. Functional comparison of paper-based immunoassays based on antibodies and engineered binding proteins. Analyst 2020; 145:2515-2519. [PMID: 32163071 DOI: 10.1039/d0an00299b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Binding protein scaffolds, such as rcSso7d, have been investigated for use in diagnostic tests; however, the functional performance of rcSso7d has not yet been studied in comparison to antibodies. Here, we assessed the analyte-binding capabilities of rcSso7d and antibodies on cellulose with samples in buffer and 100% human serum.
Collapse
Affiliation(s)
- Ki-Joo Sung
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.
| | | | | | | | | |
Collapse
|
23
|
Miller EA, Sung KJ, Kongsuphol P, Baniya S, Aw-Yong HQ, Tay V, Tan Y, Kabir FM, Pang-Yeo K, Kaspriskie IG, Sikes HD. Beyond Epitope Binning: Directed in Vitro Selection of Complementary Pairs of Binding Proteins. ACS COMBINATORIAL SCIENCE 2020; 22:49-60. [PMID: 31769955 DOI: 10.1021/acscombsci.9b00176] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Many biotechnological applications require the simultaneous binding of affinity reagents to nonoverlapping target epitopes, the most prominent example being sandwich immunoassays. Typically, affinity pairs are identified via post facto functional analysis of clones that were not selected for complementarity. Here, we developed the Rapid Affinity Pair Identification via Directed Selection (RAPIDS) process, which enables the efficient identification of affinity reagents that function together as complementary pairs, from in vitro libraries of ∼109 variants. We used RAPIDS to develop highly specific affinity pairs against biomarkers of tuberculosis, Zika virus, and sepsis. Without additional trial-and-error screening, these affinity pairs exhibited utility in multiple assay formats. The RAPIDS process applies selective pressure to hundreds of thousands of potential affinity pairs to efficiently identify complementary pairs that bind to separate epitopes without binding to one another or nontargets, yielding diagnostic assays that are sensitive and specific by design.
Collapse
Affiliation(s)
- Eric A. Miller
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Ki-Joo Sung
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Patthara Kongsuphol
- Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 CREATE Way, Singapore 138602
| | - Subha Baniya
- Department of Biochemistry, Wellesley College, Wellesley, Massachusetts 02481, United States
| | - Hui Qi Aw-Yong
- Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 CREATE Way, Singapore 138602
| | - Vivian Tay
- Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 CREATE Way, Singapore 138602
| | - Yuxuan Tan
- Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 CREATE Way, Singapore 138602
| | - Farah M. Kabir
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Karl Pang-Yeo
- Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 CREATE Way, Singapore 138602
| | - Isabel G. Kaspriskie
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Hadley D. Sikes
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 CREATE Way, Singapore 138602
| |
Collapse
|
24
|
El Hage K, Zoete V. Strong Enrichment of Aromatic and Sulfur-Containing Residues in Ligand-Protein Binding Sites. J Chem Inf Model 2019; 59:4921-4928. [PMID: 31661621 DOI: 10.1021/acs.jcim.9b00582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
While certain residues have clear involvement in determining the 3D structure of a macromolecule because they affect the folding topology or the overall protein stability, the role of different residues in ligand accommodation and binding has attracted less attention. On the basis of the assumption that drug-binding sites on target molecules have specific amino acid compositions, the incidence of each standard amino acid at the binding sites of small molecules and their correlations are calculated for an unprecedented large set of high-quality X-ray structures. Results show, for the first time, strong and highly correlated enrichments of aromatic and sulfur-containing residues, which play an important role in ligand binding and shape the nature of the chemical interactions.
Collapse
Affiliation(s)
- Krystel El Hage
- Computer-Aided Molecular Engineering Group, Department of Fundamental Oncology , University of Lausanne , Ludwig Lausanne Branch, Route de la Corniche 9A , 1066 Epalinges , Switzerland
| | - Vincent Zoete
- Computer-Aided Molecular Engineering Group, Department of Fundamental Oncology , University of Lausanne , Ludwig Lausanne Branch, Route de la Corniche 9A , 1066 Epalinges , Switzerland.,Molecular Modeling Group , SIB Swiss Institute of Bioinformatics , Quartier UNIL-Sorge, Bâtiment Amphipole , 1015 Lausanne , Switzerland
| |
Collapse
|
25
|
Zorzi A, Linciano S, Angelini A. Non-covalent albumin-binding ligands for extending the circulating half-life of small biotherapeutics. MEDCHEMCOMM 2019; 10:1068-1081. [PMID: 31391879 PMCID: PMC6644573 DOI: 10.1039/c9md00018f] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 06/01/2019] [Indexed: 12/13/2022]
Abstract
Peptides and small protein scaffolds are gaining increasing interest as therapeutics. Similarly to full-length antibodies, they can bind a target with a high binding affinity and specificity while remaining small enough to diffuse into tissues. However, despite their numerous advantages, small biotherapeutics often suffer from a relatively short circulating half-life, thus requiring frequent applications that ultimately restrict their ease of use and user compliance. To overcome this limitation, a large variety of half-life extension strategies have been developed in the last decades. Linkage to ligands that non-covalently bind to albumin, the most abundant serum protein with a circulating half-life of ∼19 days in humans, represents one of the most successful approaches for the generation of long-lasting biotherapeutics with improved pharmacokinetic properties and superior efficacy in the clinic.
Collapse
Affiliation(s)
- Alessandro Zorzi
- Institute of Chemical Sciences and Engineering , School of Basic Sciences , Ecole Polytechnique Fédérale de Lausanne (EPFL) , Lausanne CH-1015 , Switzerland
| | - Sara Linciano
- Department of Molecular Sciences and Nanosystems , Ca' Foscari University of Venice , Via Torino 155 , Venezia Mestre 30172 , Italy
| | - Alessandro Angelini
- Department of Molecular Sciences and Nanosystems , Ca' Foscari University of Venice , Via Torino 155 , Venezia Mestre 30172 , Italy
- European Centre for Living Technologies (ECLT) , San Marco 2940 , Venice 30124 , Italy .
| |
Collapse
|
26
|
Zhang Q, Zeininger L, Sung KJ, Miller EA, Yoshinaga K, Sikes HD, Swager TM. Emulsion Agglutination Assay for the Detection of Protein-Protein Interactions: An Optical Sensor for Zika Virus. ACS Sens 2019; 4:180-184. [PMID: 30624045 DOI: 10.1021/acssensors.8b01202] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A new class of Janus emulsion agglutination assay is reported for the detection of interfacial protein-protein interactions. Janus emulsion droplets are functionalized with a thermally stable, antigen binding protein rcSso7d variant (rcSso7d-ZNS1) for the detection of Zika NS1 protein. The emulsion droplets containing fluorescent dyes in their hydrocarbon and fluorocarbon phases intensify the intrinsic optical signal with the emission intensity ratio, which can be detected by a simple optical fiber. This assay provides an opportunity for the in-field detection of Zika virus and other pathogens with a stable, inexpensive, and convenient device.
Collapse
Affiliation(s)
- Qifan Zhang
- Department of Chemistry and Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Lukas Zeininger
- Department of Chemistry and Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Ki-Joo Sung
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Eric A. Miller
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Kosuke Yoshinaga
- Department of Chemistry and Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Hadley D. Sikes
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Timothy M. Swager
- Department of Chemistry and Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
27
|
Paloni JM, Miller EA, Sikes HD, Olsen BD. Improved Ordering in Low Molecular Weight Protein-Polymer Conjugates Through Oligomerization of the Protein Block. Biomacromolecules 2018; 19:3814-3824. [PMID: 30132651 DOI: 10.1021/acs.biomac.8b00928] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The self-assembly of protein-polymer conjugates incorporating oligomers of a small, engineered high-affinity binding protein, rcSso7d.SA, is studied to determine the effect of protein oligomerization on nanoscale ordering. Oligomerization enables a systematic increase in the protein molar mass without changing its overall folded structure, leading to a higher driving force for self-assembly into well-ordered structures. Though conjugates of monomeric rcSso7d.SA are found to only exist in disordered states, oligomers of this protein linked to a poly( N-isopropylacrylamide) (PNIPAM) block self-assemble into lamellar nanostructures. Conjugates of trimeric and tetrameric rcSso7d.SA are observed to produce the strongest ordering in concentrated solution, displaying birefringent lamellae at concentrations as low as 40 wt %. In highly concentrated solution, the oligomeric rcSso7d.SA-PNIPAM block copolymers exhibit ordering and domain spacing trends atypical from that of most block copolymers. Fluorescent binding assays indicate that oligomerized protein blocks retain binding functionality and exhibit limits of detection up to three times lower than that of surface-immobilized protein sensors. Therefore, oligomerization of the protein block in these block copolymers serves as an effective method to improve both nanoscale ordering and biosensing capabilities.
Collapse
Affiliation(s)
- Justin M Paloni
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Eric A Miller
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Hadley D Sikes
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Bradley D Olsen
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| |
Collapse
|
28
|
Abstract
Peptide secondary and tertiary structure motifs frequently serve as inspiration for the development of protein-protein interaction (PPI) inhibitors. While a wide variety of strategies have been used to stabilize or imitate α-helices, similar strategies for β-sheet stabilization are more limited. Synthetic scaffolds that stabilize reverse turns and cross-strand interactions have provided important insights into β-sheet stability and folding. However, these templates occupy regions of the β-sheet that might impact the β-sheet's ability to bind at a PPI interface. Here, we present the hydrogen bond surrogate (HBS) approach for stabilization of β-hairpin peptides. The HBS linkage replaces a cross-strand hydrogen bond with a covalent linkage, conferring significant conformational and proteolytic resistance. Importantly, this approach introduces the stabilizing linkage in the buried β-sheet interior, retains all side chains for further functionalization, and allows efficient solid-phase macrocyclization. We anticipate that HBS stabilization of PPI β-sheets will enhance the development of β-sheet PPI inhibitors and expand the repertoire of druggable PPIs.
Collapse
Affiliation(s)
- Nicholas Sawyer
- Department of Chemistry, New York University, 100 Washington Square East, New York, New York 10003, United States
| | - Paramjit S. Arora
- Department of Chemistry, New York University, 100 Washington Square East, New York, New York 10003, United States
| |
Collapse
|
29
|
Kruziki MA, Sarma V, Hackel BJ. Constrained Combinatorial Libraries of Gp2 Proteins Enhance Discovery of PD-L1 Binders. ACS COMBINATORIAL SCIENCE 2018; 20:423-435. [PMID: 29799714 DOI: 10.1021/acscombsci.8b00010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Engineered protein ligands are used for molecular therapy, diagnostics, and industrial biotechnology. The Gp2 domain is a 45-amino acid scaffold that has been evolved for specific, high-affinity binding to multiple targets by diversification of two solvent-exposed loops. Inspired by sitewise enrichment of select amino acids, including cysteine pairs, in earlier Gp2 discovery campaigns, we hypothesized that the breadth and efficiency of de novo Gp2 discovery will be aided by sitewise amino acid constraint within combinatorial library design. We systematically constructed eight libraries and comparatively evaluated their efficacy for binder discovery via yeast display against a panel of targets. Conservation of a cysteine pair at the termini of the first diversified paratope loop increased binder discovery 16-fold ( p < 0.001). Yet two other libraries with conserved cysteine pairs, within the second loop or an interloop pair, did not aid discovery thereby indicating site-specific impact. Via a yeast display protease resistance assay, Gp2 variants from the loop one cysteine pair library were 3.3 ± 2.1-fold ( p = 0.005) more stable than nonconstrained variants. Sitewise constraint of noncysteine residues-guided by previously evolved binders, natural Gp2 homology, computed stability, and structural analysis-did not aid discovery. A panel of binders to programmed death ligand 1 (PD-L1), a key target in cancer immunotherapy, were discovered from the loop 1 cysteine constraint library. Affinity maturation via loop walking resulted in strong, specific cellular PD-L1 affinity ( Kd = 6-9 nM).
Collapse
Affiliation(s)
- Max A. Kruziki
- University of Minnesota—Twin Cities, Department of Chemical Engineering and Materials Science, 421 Washington Avenue Southeast, Minneapolis, Minnesota 55455, United States
| | - Vidur Sarma
- University of Minnesota—Twin Cities, Department of Chemical Engineering and Materials Science, 421 Washington Avenue Southeast, Minneapolis, Minnesota 55455, United States
| | - Benjamin J. Hackel
- University of Minnesota—Twin Cities, Department of Chemical Engineering and Materials Science, 421 Washington Avenue Southeast, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
30
|
Case BA, Kruziki MA, Johnson SM, Hackel BJ. Engineered Charge Redistribution of Gp2 Proteins through Guided Diversity for Improved PET Imaging of Epidermal Growth Factor Receptor. Bioconjug Chem 2018; 29:1646-1658. [PMID: 29579383 PMCID: PMC6051758 DOI: 10.1021/acs.bioconjchem.8b00144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The Gp2 domain is a protein scaffold for synthetic ligand engineering. However, the native protein function results in a heterogeneous distribution of charge on the conserved surface, which may hinder further development and utility. We aim to modulate charge, without diminishing function, which is challenging in small proteins where each mutation is a significant fraction of protein structure. We constructed rationally guided combinatorial libraries with charge-neutralizing or charge-flipping mutations and sorted them, via yeast display and flow cytometry, for stability and target binding. Deep sequencing of functional variants revealed effective mutations both in clone-dependent contexts and broadly across binders to epidermal growth factor receptor (EGFR), insulin receptor, and immunoglobulin G. Functional mutants averaged 4.3 charge neutralizing mutations per domain while maintaining net negative charge. We evolved an EGFR-targeted Gp2 mutant that reduced charge density by 33%, maintained net charge, and improved charge distribution homogeneity while elevating thermal stability ( Tm = 87 ± 1 °C), improving binding specificity, and maintaining affinity ( Kd = 8.8 ± 0.6 nM). This molecule was conjugated with 1,4,7-triazacyclononane,1-glutaric acid-4,7-acetic acid for 64Cu chelation and evaluated for physiological distribution in mice with xenografted A431 (EGFRhigh) and MDA-MB-435 (EGFRlow) tumors. Excised tissue gamma counting and positron emission tomography/computed tomography imaging revealed good EGFRhigh tumor signal (4.7 ± 0.5%ID/g) at 2 h post-injection and molecular specificity evidenced by low uptake in EGFRlow tumors (0.6 ± 0.1%ID/g, significantly lower than for non-charge-modified Gp2, p = 0.01). These results provide charge mutations for an improved Gp2 framework, validate an effective approach to charge engineering, and advance performance of physiological EGFR targeting for molecular imaging.
Collapse
Affiliation(s)
- Brett A. Case
- University of Minnesota – Twin Cities, Department of Chemical Engineering and Materials Science, 421 Washington Avenue SE, Minneapolis, MN 55455
| | - Max A. Kruziki
- University of Minnesota – Twin Cities, Department of Chemical Engineering and Materials Science, 421 Washington Avenue SE, Minneapolis, MN 55455
| | - Sadie M. Johnson
- University of Minnesota – Twin Cities, Department of Chemical Engineering and Materials Science, 421 Washington Avenue SE, Minneapolis, MN 55455
| | - Benjamin J. Hackel
- University of Minnesota – Twin Cities, Department of Chemical Engineering and Materials Science, 421 Washington Avenue SE, Minneapolis, MN 55455
| |
Collapse
|
31
|
Kauke MJ, Tisdale AW, Kelly RL, Braun CJ, Hemann MT, Wittrup KD. A Raf-Competitive K-Ras Binder Can Fail to Functionally Antagonize Signaling. Mol Cancer Ther 2018; 17:1773-1780. [PMID: 29720559 DOI: 10.1158/1535-7163.mct-17-0645] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 11/15/2017] [Accepted: 04/23/2018] [Indexed: 11/16/2022]
Abstract
Mutated in approximately 30% of human cancers, Ras GTPases are the most common drivers of oncogenesis and render tumors unresponsive to many standard therapies. Despite decades of research, no drugs directly targeting Ras are currently available. We have previously characterized a small protein antagonist of K-Ras, R11.1.6, and demonstrated its direct competition with Raf for Ras binding. Here we evaluate the effects of R11.1.6 on Ras signaling and cellular proliferation in a panel of human cancer cell lines. Through lentiviral transduction, we generated cell lines that constitutively or through induction with doxycycline express R11.1.6 or a control protein YW1 and show specific binding by R11.1.6 to endogenous Ras through microscopy and co-immunoprecipitation experiments. Genetically encoded intracellular expression of this high-affinity Ras antagonist, however, fails to measurably disrupt signaling through either the MAPK or PI3K pathway. Consistently, cellular proliferation was unaffected as well. To understand this lack of signaling inhibition, we quantified the number of molecules of R11.1.6 expressed by the inducible cell lines and developed a simple mathematical model describing the competitive binding of Ras by R11.1.6 and Raf. This model supports a potential mechanism for the lack of biological effects that we observed, suggesting stoichiometric and thermodynamic barriers that should be overcome in pharmacologic efforts to directly compete with downstream effector proteins localized to membranes at very high effective concentrations. Mol Cancer Ther; 17(8); 1773-80. ©2018 AACR.
Collapse
Affiliation(s)
- Monique J Kauke
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Alison W Tisdale
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Ryan L Kelly
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Christian J Braun
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Michael T Hemann
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - K Dane Wittrup
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts. .,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
32
|
Wang M, Zhu D, Zhu J, Nussinov R, Ma B. Local and global anatomy of antibody-protein antigen recognition. J Mol Recognit 2018; 31:e2693. [PMID: 29218757 PMCID: PMC5903993 DOI: 10.1002/jmr.2693] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 09/13/2017] [Accepted: 11/08/2017] [Indexed: 12/15/2022]
Abstract
Deciphering antibody-protein antigen recognition is of fundamental and practical significance. We constructed an antibody structural dataset, partitioned it into human and murine subgroups, and compared it with nonantibody protein-protein complexes. We investigated the physicochemical properties of regions on and away from the antibody-antigen interfaces, including net charge, overall antibody charge distributions, and their potential role in antigen interaction. We observed that amino acid preference in antibody-protein antigen recognition is entropy driven, with residues having low side-chain entropy appearing to compensate for the high backbone entropy in interaction with protein antigens. Antibodies prefer charged and polar antigen residues and bridging water molecules. They also prefer positive net charge, presumably to promote interaction with negatively charged protein antigens, which are common in proteomes. Antibody-antigen interfaces have large percentages of Tyr, Ser, and Asp, but little Lys. Electrostatic and hydrophobic interactions in the Ag binding sites might be coupled with Fab domains through organized charge and residue distributions away from the binding interfaces. Here we describe some features of antibody-antigen interfaces and of Fab domains as compared with nonantibody protein-protein interactions. The distributions of interface residues in human and murine antibodies do not differ significantly. Overall, our results provide not only a local but also a global anatomy of antibody structures.
Collapse
Affiliation(s)
- Meryl Wang
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702
| | - David Zhu
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702
| | - Jianwei Zhu
- School of Pharmacy, Shanghai Jiao Tong University, 800 DongChuan Road, Shanghai 200240, China
| | - Ruth Nussinov
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702
- Sackler Inst. of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Buyong Ma
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702
| |
Collapse
|
33
|
Miller EA, Baniya S, Osorio D, Al Maalouf YJ, Sikes HD. Paper-based diagnostics in the antigen-depletion regime: High-density immobilization of rcSso7d-cellulose-binding domain fusion proteins for efficient target capture. Biosens Bioelectron 2018; 102:456-463. [PMID: 29182928 PMCID: PMC5983361 DOI: 10.1016/j.bios.2017.11.050] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/17/2017] [Accepted: 11/16/2017] [Indexed: 11/28/2022]
Abstract
In this work, we report the development of a general strategy for enhancing the efficiency of target capture in immunoassays, using a bifunctional fusion protein construct which incorporates a substrate-anchoring moiety for the high-abundance immobilization of an antigen-binding domain. This approach was informed by the development of a pseudo first-order rate constant model, and tested in a paper-based assay format using a fusion construct consisting of an rcSso7d binding module and a cellulose-binding domain. These rcSso7d-CBD fusion proteins were solubly expressed and purified from bacteria in high molar yields, and enable oriented, high-density adsorption of the rcSso7d binding species to unmodified cellulose within a 30-second incubation period. These findings were validated using two distinct, antigen-specific rcSso7d variants, which were isolated from a yeast surface display library via flow cytometry. Up to 1.6 micromoles of rcSso7d-CBD was found to adsorb per gram of cellulose, yielding a volume-averaged binder concentration of up to 760μM within the resulting active material. At this molar abundance, the target antigen is captured from solution with nearly 100% efficiency, maximizing the attainable sensitivity for any given diagnostic system.
Collapse
Affiliation(s)
- Eric A Miller
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Subha Baniya
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Daniel Osorio
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Yara Jabbour Al Maalouf
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Hadley D Sikes
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| |
Collapse
|
34
|
Case BA, Kruziki MA, Stern LA, Hackel BJ. Evaluation of affibody charge modification identified by synthetic consensus design in molecular PET imaging of epidermal growth factor receptor. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2018; 3:171-182. [PMID: 31467687 PMCID: PMC6715147 DOI: 10.1039/c7me00095b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Tumor overexpression of epidermal growth factor receptor (EGFR) correlates to therapeutic response in select patient populations. Thus, molecular positron emission tomography (PET) imaging of EGFR could stratify responders versus non-responders. We previously demonstrated effectiveness of a "synthetic consensus" design principle to identify six neutralizing mutations within a 58-amino acid EGFR-targeted affibody domain. Herein, we extend the approach to identify additional neutralized variants that vary net charge from -2 to either -4 or +4 while retaining high affinity (1.6 ± 1.2 nM and 2.5 ± 0.7 nM), specific binding to EGFR, secondary structure, and stability (Tm = 68 °C and 59 °C). We radiolabeled the resultant collection of five charge variants with 64Cu and evaluated PET imaging performance in murine models with subcutaneously xenografted EGFRhigh and EGFRlow tumors. All variants exhibited good EGFRhigh tumor imaging as early as 1 h, with EA35S (+3/-5) achieving 7.7 ± 1.4 %ID/g tumor at 4 h with 1.5 ± 0.3%ID/g EGFRlow tumor, 34 ± 5 tumor:muscle and 12 ± 3 tumor:blood ratios. The positively charged EA62S mutant (+6/-2) exhibited 2.2-3.3-fold higher liver signal than the other variants (p<0.01). The EA68 variant with higher charge density was more stable to human and mouse serum than neutralized variants. In a comparison of radiometal chelators, 1,4,7-triazacyclononane,1-glutaric acid-4,7-acetic acid (NODAGA) exhibited superior physiological specificity to 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA). In total, these studies comparatively evaluated a set of EGFR-targeted affibodies varying in net charge and charge density, which revealed functional variations that are useful in engineering an ideal probe for translational studies.
Collapse
Affiliation(s)
- Brett A Case
- Department of Chemical Engineering and Materials Science, University of Minnesota - Twin Cities, Minneapolis, MN 55455
| | - Max A Kruziki
- Department of Chemical Engineering and Materials Science, University of Minnesota - Twin Cities, Minneapolis, MN 55455
| | - Lawrence A Stern
- Department of Chemical Engineering and Materials Science, University of Minnesota - Twin Cities, Minneapolis, MN 55455
| | - Benjamin J Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota - Twin Cities, Minneapolis, MN 55455
| |
Collapse
|
35
|
Könning D, Kolmar H. Beyond antibody engineering: directed evolution of alternative binding scaffolds and enzymes using yeast surface display. Microb Cell Fact 2018; 17:32. [PMID: 29482656 PMCID: PMC6389260 DOI: 10.1186/s12934-018-0881-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/22/2018] [Indexed: 01/08/2023] Open
Abstract
Pioneered exactly 20 years ago, yeast surface display (YSD) continues to take a major role in protein engineering among the high-throughput display methodologies that have been developed to date. The classical yeast display technology relies on tethering an engineered protein to the cell wall by genetic fusion to one subunit of a dimeric yeast-mating agglutination receptor complex. This method enables an efficient genotype-phenotype linkage while exploiting the benefits of a eukaryotic expression machinery. Over the past two decades, a plethora of protein engineering efforts encompassing conventional antibody Fab and scFv fragments have been reported. In this review, we will focus on the versatility of YSD beyond conventional antibody engineering and, instead, place the focus on alternative scaffold proteins and enzymes which have successfully been tailored for purpose with regard to improving binding, activity or specificity.
Collapse
Affiliation(s)
- Doreen Könning
- Antibody-Drug Conjugates and Targeted NBE Therapeutics, Merck KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, 64287 Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, 64287 Darmstadt, Germany
| |
Collapse
|
36
|
Cruz-Teran CA, Tiruthani K, Mischler A, Rao BM. Inefficient Ribosomal Skipping Enables Simultaneous Secretion and Display of Proteins in Saccharomyces cerevisiae. ACS Synth Biol 2017; 6:2096-2107. [PMID: 28805373 PMCID: PMC5905331 DOI: 10.1021/acssynbio.7b00144] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The need for recombinant expression of soluble protein slows the validation of engineered proteins isolated from combinatorial libraries and limits the number of protein variants evaluated. To overcome this bottleneck, we describe a system for simultaneous cell surface display and soluble secretion of proteins in Saccharomyces cerevisiae based on inefficient ribosomal skipping. Ribosomal skipping mediated by "self-cleaving" 2A peptides produces two proteins from a single open reading frame. Incorporation of the F2A peptide sequence-with ∼50% efficiency of ribosomal skipping-between the protein of interest and the yeast cell wall protein Aga2 results in simultaneous expression of both the solubly secreted protein and the protein-Aga2 fusion that is tethered to the yeast cell surface. We show that binding proteins derived from the Sso7d scaffold and the homodimeric enzyme glucose oxidase can be simultaneously secreted solubly and expressed as yeast cell surface fusions using the F2A-based system. Furthermore, a combinatorial library of Sso7d mutants can be screened to isolate binders with higher affinity for a model target (lysozyme), and the pool of higher affinity binders can be characterized in soluble form. Significantly, we show that both N- and C-terminal fusions to Aga2 can be simultaneously secreted solubly and displayed on the cell surface; this is particularly advantageous because protein functionality can be affected by the specific position of Aga2 in the protein fusion. We expect that the F2A-based yeast surface display and secretion system will be a useful tool for protein engineering and enable efficient characterization of individual clones isolated from combinatorial libraries.
Collapse
Affiliation(s)
| | | | - Adam Mischler
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC
| | - Balaji M. Rao
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC
| |
Collapse
|
37
|
Yang NJ, Kauke MJ, Sun F, Yang LF, Maass KF, Traxlmayr MW, Yu Y, Xu Y, Langer RS, Anderson DG, Wittrup KD. Cytosolic delivery of siRNA by ultra-high affinity dsRNA binding proteins. Nucleic Acids Res 2017. [PMID: 28641400 PMCID: PMC5570165 DOI: 10.1093/nar/gkx546] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Protein-based methods of siRNA delivery are capable of uniquely specific targeting, but are limited by technical challenges such as low potency or poor biophysical properties. Here, we engineered a series of ultra-high affinity siRNA binders based on the viral protein p19 and developed them into siRNA carriers targeted to the epidermal growth factor receptor (EGFR). Combined in trans with a previously described endosome-disrupting agent composed of the pore-forming protein Perfringolysin O (PFO), potent silencing was achieved in vitro with no detectable cytotoxicity. Despite concerns that excessively strong siRNA binding could prevent the discharge of siRNA from its carrier, higher affinity continually led to stronger silencing. We found that this improvement was due to both increased uptake of siRNA into the cell and improved pharmacodynamics inside the cell. Mathematical modeling predicted the existence of an affinity optimum that maximizes silencing, after which siRNA sequestration decreases potency. Our study characterizing the affinity dependence of silencing suggests that siRNA-carrier affinity can significantly affect the intracellular fate of siRNA and may serve as a handle for improving the efficiency of delivery. The two-agent delivery system presented here possesses notable biophysical properties and potency, and provide a platform for the cytosolic delivery of nucleic acids.
Collapse
Affiliation(s)
- Nicole J Yang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Monique J Kauke
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Fangdi Sun
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lucy F Yang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Katie F Maass
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael W Traxlmayr
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yao Yu
- Protein Analytics, Adimab LLC, Lebanon, NH 03766, USA
| | - Yingda Xu
- Protein Analytics, Adimab LLC, Lebanon, NH 03766, USA
| | - Robert S Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel G Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - K Dane Wittrup
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
38
|
Identification and characterization of a novel Sso7d scaffold-based binder against Notch1. Sci Rep 2017; 7:12021. [PMID: 28931897 PMCID: PMC5607287 DOI: 10.1038/s41598-017-12246-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 09/06/2017] [Indexed: 12/18/2022] Open
Abstract
Notch signaling has important functions in regulating cell growth and development, misregulation of which has been implicated in various cancers. Monoclonal antibodies (mAbs) targeting Notch protein activity have already moved into clinical trials. However due to the limitations associated with cost and productivity of mAbs, there has been a surge in the development of complementary approaches that are based on non-antibody scaffolds. Non-antibody scaffolds are small proteins that are stable and can be engineered to develop high-affinity binders against specific targets of interest. Here we describe the isolation and characterization of a novel Notch1-binding protein, N9, obtained by screening of a combinatorial library based on the ultra-stable Sso7d scaffold. N9 targets the extracellular EGF-like repeats (ELR) 11–13 in Notch1, and therefore serves as a competitive inhibitor for Notch ligands to decrease expression of Notch target genes. We demonstrate that N9 recognizes surface expression of Notch1 on the plasma membrane and binds preferentially to cell lines misexpressing Notch1. Although N9 was selected against Notch1, we also observe cross-reactivity against other Notch receptors, including Notch2/3. Finally, we demonstrate that N9 inhibits proliferation and generation of tumorspheres in Notch expressing cancer cell lines, suggesting its potential as a therapeutic agent in Notch-associated malignancies.
Collapse
|
39
|
Kelly RL, Geoghegan JC, Feldman J, Jain T, Kauke M, Le D, Zhao J, Wittrup KD. Chaperone proteins as single component reagents to assess antibody nonspecificity. MAbs 2017; 9:1036-1040. [PMID: 28745541 PMCID: PMC5627595 DOI: 10.1080/19420862.2017.1356529] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Early stage assays that evaluate monoclonal antibody drug-like properties serve as valuable tools for selection of lead candidates. One liability for clinical development, off-target reactivity, is often assessed by binding to a mixture or panel of noncognate proteins. While robust, these mixes are often ill-defined, and can suffer from issues such as lot-to-lot variability. In this study, we discovered in immunoprecipitation experiments that certain chaperones are present in one of these mixtures;we then explored the use of recombinant chaperone proteins as well-characterized agents to predict antibody nonspecificity. Antibody binding to the heat shock proteins HSP70, HSP90, or trigger factor all served as predictors of cross-interaction propensity, with HSP90 providing the greatest ability to predict antibody clearance rates in mouse. Individual chaperone binding correlates surprisingly closely with binding to complex cell extracts, with the exception of a few “false negatives” (assuming a complex cell extract as the “true” value). As defined reagents, these chaperone reagents present advantages for high throughput assays of nonspecificity.
Collapse
Affiliation(s)
- Ryan L Kelly
- a Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , USA
| | | | | | | | - Monique Kauke
- b Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA U.S.A
| | - Doris Le
- b Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA U.S.A
| | - Jessie Zhao
- b Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA U.S.A
| | - K Dane Wittrup
- a Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , USA.,b Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA U.S.A
| |
Collapse
|
40
|
Kauke MJ, Traxlmayr MW, Parker JA, Kiefer JD, Knihtila R, McGee J, Verdine G, Mattos C, Wittrup KD. An engineered protein antagonist of K-Ras/B-Raf interaction. Sci Rep 2017; 7:5831. [PMID: 28724936 PMCID: PMC5517481 DOI: 10.1038/s41598-017-05889-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/05/2017] [Indexed: 12/31/2022] Open
Abstract
Ras is at the hub of signal transduction pathways controlling cell proliferation and survival. Its mutants, present in about 30% of human cancers, are major drivers of oncogenesis and render tumors unresponsive to standard therapies. Here we report the engineering of a protein scaffold for preferential binding to K-Ras G12D. This is the first reported inhibitor to achieve nanomolar affinity while exhibiting specificity for mutant over wild type (WT) K-Ras. Crystal structures of the protein R11.1.6 in complex with K-Ras WT and K-Ras G12D offer insight into the structural basis for specificity, highlighting differences in the switch I conformation as the major defining element in the higher affinity interaction. R11.1.6 directly blocks interaction with Raf and reduces signaling through the Raf/MEK/ERK pathway. Our results support greater consideration of the state of switch I and provide a novel tool to study Ras biology. Most importantly, this work makes an unprecedented contribution to Ras research in inhibitor development strategy by revealing details of a targetable binding surface. Unlike the polar interfaces found for Ras/effector interactions, the K-Ras/R11.1.6 complex reveals an extensive hydrophobic interface that can serve as a template to advance the development of high affinity, non-covalent inhibitors of K-Ras oncogenic mutants.
Collapse
Affiliation(s)
- Monique J Kauke
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Michael W Traxlmayr
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Jillian A Parker
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, 02115, USA
| | - Jonathan D Kiefer
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Ryan Knihtila
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, 02115, USA
| | - John McGee
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Greg Verdine
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Carla Mattos
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, 02115, USA
| | - K Dane Wittrup
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA. .,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA. .,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|