Increased Tumor Penetration of Single-Domain Antibody Drug

Total Page:16

File Type:pdf, Size:1020Kb

Increased Tumor Penetration of Single-Domain Antibody Drug Author Manuscript Published OnlineFirst on January 15, 2020; DOI: 10.1158/0008-5472.CAN-19-2295 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Increased Tumor Penetration of Single-Domain Antibody Drug Conjugates Improves In Vivo Efficacy in Prostate Cancer Models Ian Nessler1, Eshita Khera1, Steven Vance2, Anna Kopp1, Qifeng Qiu3, Thomas A. Keating3, Adnan O. Abu-Yousif4, Thomas Sandal2, James Legg2, Lorraine Thompson2, Normann Goodwin2, and Greg M. Thurber1,5 1 – Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 2 – Crescendo Biologics, Cambridge, United Kingdom 3 – ImmunoGen, Waltham, MA, 02451 4 – Takeda Pharmaceuticals, Cambridge, MA, 02139 5 – Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109 Statement of Significance A mechanistic study of protein-drug conjugates demonstrates that a lower potency compound is more effective in vivo than other agents with equal tumor uptake due to improved tissue penetration and cellular distribution. Conflict of Interest SV, TS, JL, LT, and NG were employed by Crescendo, QQ and TAK were employed by Immunogen, and AA was employed by Takeda during the study. GMT sits on the Scientific Advisory Board of Advanced Proteome Therapeutics. Running Title: Tumor penetration drives efficacy of protein-drug conjugates Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 15, 2020; DOI: 10.1158/0008-5472.CAN-19-2295 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Abstract Targeted delivery of chemotherapeutics aims to increase efficacy and lower toxicity by concentrating drugs at the site-of-action, a method embodied by the seven current FDA approved antibody-drug conjugates (ADCs). However, a variety of pharmacokinetic challenges result in relatively narrow therapeutic windows for these agents, hampering the development of new drugs. Here, we use a series of Prostate-Specific Membrane Antigen (PSMA)-binding single- domain (Humabody®) ADC constructs to demonstrate that tissue penetration of protein-drug conjugates plays a major role in therapeutic efficacy. Counterintuitively, a construct with lower in vitro potency resulted in higher in vivo efficacy than other protein-drug conjugates. Biodistribution data, tumor histology images, spheroid experiments, in vivo single-cell measurements, and computational results demonstrate that a smaller size and slower internalization rate enabled higher tissue penetration and more cell killing. The results also illustrate the benefits of linking an albumin binding domain to the single-domain ADCs. A construct lacking an albumin binding domain was rapidly cleared leading to lower tumor uptake (%ID/g) and decreased in vivo efficacy. In conclusion, these results provide evidence that reaching the maximum number of cells with a lethal payload dose correlates more strongly with in vivo efficacy than total tumor uptake or in vitro potency alone for these protein-drug conjugates. Computational modeling and protein engineering can be used to custom design an optimal framework for controlling internalization, clearance, and tissue penetration to maximize cell killing. Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 15, 2020; DOI: 10.1158/0008-5472.CAN-19-2295 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Introduction Antibody-drug conjugates (ADCs) have opened a new field of targeted therapeutics based on ‘hybrid’ drugs combining desirable targeting properties of biologics with the potency of small molecule cytotoxic payloads. For ADCs, the protein carrier is typically a monoclonal antibody that specifically binds to a target antigen expressed on cancer cells, increasing the delivery of the small molecule payload to the site of action in vivo. To date, seven ADCs have been FDA approved1, 2, most recently enfortumab vedotin and trastuzumab deruxtecan, with a large pipeline of >70 in clinical trials. However, one drawback of antibodies is slow tumor penetration. The tumor uptake of antibodies is limited by their extravasation rate, and they tend to penetrate only a few cell layers outside of blood vessels due to their rapid antigen-binding rate relative to intratumoral diffusion3, 4. In the clinic, unconjugated antibodies are often well tolerated, such that they can be delivered at very high doses that saturate receptors on cell layers closer to the blood vessel, enabling the antibody to diffuse farther through the tumor. However, the payload toxicity of ADCs limits the dose and frequency of administration, restricting tumor penetration depths and allowing regrowth between doses (typically given every 3 weeks for current therapies)5. Therefore, devising design and treatment strategies that increase tumor penetration may yield greater efficacy and improve clinical success rates for ADCs and other protein-drug conjugates. Many current ADCs in late-stage clinical trials are targeting receptors with high expression (e.g. TROP-2, HER2, folate receptor, and Nectin-4) where tissue penetration can play a significant role in efficacy. Previous work based on co-administration of trastuzumab with the ADC T-DM1 (ado-trastuzumab emtansine) demonstrated that higher tissue penetration could yield better efficacy in a mouse model of highly expressed HER2 positive cancer6. Based on a literature review of studies involving ADCs with a range of drug-to-antibody ratios (DAR)7, ADCs that delivered the same payload dose at a lower DAR (i.e. higher protein doses) generally yielded an increase in efficacy in animal models with moderate to high expression, likely as a result of better tissue penetration8. Although bystander effects can help mitigate transport challenges by allowing the payload to diffuse deeper into the tissue following release from the protein, analysis of literature data and predictive simulations indicated that higher tissue penetration is more efficient at improving efficacy even when using bystander payloads8. When the ADC is more uniformly distributed, the same total amount of cytotoxic payload delivered to the tumor is spread more homogeneously, so cells adjacent to vessels that receive an overdose of payload with heterogeneous ADC distribution now receive fewer payloads. Therefore, implicit in this approach is that the amount of payload delivered per cell exceeds the intrinsic potency of the payload, thereby maintaining a lethal cellular dose while increasing penetration to reach more cells. Similarly, lower target receptor expression can improve efficacy for extremely potent ADCs by enabling deeper tissue penetration. Lower receptor expression limits the amount of bound and internalized ADC for perivascular cells but increases tumor tissue penetration since penetration depth is inversely proportional to expression7, 9. Based on these and other results10, tumor tissue penetration is critical for the overall efficacy of ADC treatment and should be analyzed when optimizing protein-drug conjugates for maximum effectiveness. Besides increasing the dose or co-administering the ADC with unconjugated antibody, there are protein engineering approaches to develop scaffolds for improved tissue penetration11- 14. This may be particularly advantageous for highly expressed targets that would require very Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 15, 2020; DOI: 10.1158/0008-5472.CAN-19-2295 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. large antibody doses to saturate the tumor or for targeting metastases with varying expression levels. Reducing affinity is one method of increasing tissue penetration15. However, for large, bivalent IgGs, the intrinsic affinity of each Fab arm must be very low to penetrate tissue with 16 highly expressed antigens (e.g. 270 nM Kd) . Instead of modulating affinity, lower molecular weight proteins can penetrate tissue farther before being immobilized through target binding17 because of their faster diffusion coefficient (which is the dominant mode of transport in tumors due to elevated interstitial pressure18). Some smaller scaffolds are already being tested in the clinic, such as caplacizumab - a bivalent single-domain antibody (~30kDa) used to block platelet aggregation19, providing precedent for these formats. A major limitation for these smaller scaffolds is their rapid renal clearance11, 20, 21. However, fusion to albumin binding domains can extend plasma half-life and avoid glomerular filtration by increasing the effective molecular weight while still enabling better tissue penetration22-26. In this work we investigated the tumor tissue pharmacokinetics and efficacy of 3 different Prostate-Specific Membrane Antigen (PSMA)-binding single-domain antibody (Humabody) drug conjugates and an IgG drug conjugate (referred to collectively as antibody drug conjugates or simply antibodies/antibody constructs if no drug is present). Humabodies are fully human, single heavy chain variable (VH) domains generated using a proprietary transgenic mouse platform27, 28 (Crescendo Biologics). The mice lack murine heavy chain, light chain, and light
Recommended publications
  • Strategies and Challenges for the Next Generation of Therapeutic Antibodies
    FOCUS ON THERAPEUTIC ANTIBODIES PERSPECTIVES ‘validated targets’, either because prior anti- TIMELINE bodies have clearly shown proof of activity in humans (first-generation approved anti- Strategies and challenges for the bodies on the market for clinically validated targets) or because a vast literature exists next generation of therapeutic on the importance of these targets for the disease mechanism in both in vitro and in vivo pharmacological models (experi- antibodies mental validation; although this does not necessarily equate to clinical validation). Alain Beck, Thierry Wurch, Christian Bailly and Nathalie Corvaia Basically, the strategy consists of develop- ing new generations of antibodies specific Abstract | Antibodies and related products are the fastest growing class of for the same antigens but targeting other therapeutic agents. By analysing the regulatory approvals of IgG-based epitopes and/or triggering different mecha- biotherapeutic agents in the past 10 years, we can gain insights into the successful nisms of action (second- or third-generation strategies used by pharmaceutical companies so far to bring innovative drugs to antibodies, as discussed below) or even the market. Many challenges will have to be faced in the next decade to bring specific for the same epitopes but with only one improved property (‘me better’ antibod- more efficient and affordable antibody-based drugs to the clinic. Here, we ies). This validated approach has a high discuss strategies to select the best therapeutic antigen targets, to optimize the probability of success, but there are many structure of IgG antibodies and to design related or new structures with groups working on this class of target pro- additional functions.
    [Show full text]
  • 2010.01) C07k 14/74 (2006.01
    ( (51) International Patent Classification: su 215 123 (CN). LIN, Yanni; F2, Building B20, Sangt- C07K 19/00 (2006.0 1) C12N 5/0783 (20 10.01) ian Street 218, Suzhou Industrial Park, Suzhou, Jiangsu C07K 14/74 (2006.01) A61K 35/1 7 (2015.01) 215123 (CN). ZHENG, Xiaocui; F2, Building B20, Sangt- C12N 15/62 (2006.01) A61P 35/00 (2006.01) ian Street 218, Suzhou Industrial Park, Suzhou, Jiangsu 215123 (CN). KONG, Hongmei; F2, Building B20, Sangt- (21) International Application Number: ian Street 218, Suzhou Industrial Park, Suzhou, Jiangsu PCT/ CN2020/ 1081 16 215123 (CN). WANG, Wenbo; F2, Building B20, Sangtian (22) International Filing Date: Street 218, Suzhou Industrial Park, Suzhou, Jiangsu 215 123 10 August 2020 (10.08.2020) (CN). FENG, Aihua; F2, Building B20, Sangtian Street 218, Suzhou Industrial Park, Suzhou, Jiangsu 215 123 (CN). (25) Filing Language: English (74) Agent: J. Z. M. C. PATENT AND TRADEMARK LAW (26) Publication Language: English OFFICE (GENERAL PARTNERSHIP); YU, Mingwei, (30) Priority Data: Room 5022, No. 335, Guo Ding Road, Yang Pu District, 201910746355.8 13 August 2019 (13.08.2019) CN Shanghai 200433 (CN). PCT/ CN20 19/124321 (81) Designated States (unless otherwise indicated, for every 10 December 2019 (10. 12.2019) CN kind of national protection av ailable) . AE, AG, AL, AM, (71) Applicant: CURE GENETICS CO., LTD. [CN/CN]; AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, BZ, Biobay A4-5 10, Xinghu Street 218, Suzhou Industrial Park, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DJ, DK, DM, DO, Suzhou, Jiangsu 215 123 (CN).
    [Show full text]
  • EURL ECVAM Recommendation on Non-Animal-Derived Antibodies
    EURL ECVAM Recommendation on Non-Animal-Derived Antibodies EUR 30185 EN Joint Research Centre This publication is a Science for Policy report by the Joint Research Centre (JRC), the European Commission’s science and knowledge service. It aims to provide evidence-based scientific support to the European policymaking process. The scientific output expressed does not imply a policy position of the European Commission. Neither the European Commission nor any person acting on behalf of the Commission is responsible for the use that might be made of this publication. For information on the methodology and quality underlying the data used in this publication for which the source is neither Eurostat nor other Commission services, users should contact the referenced source. EURL ECVAM Recommendations The aim of a EURL ECVAM Recommendation is to provide the views of the EU Reference Laboratory for alternatives to animal testing (EURL ECVAM) on the scientific validity of alternative test methods, to advise on possible applications and implications, and to suggest follow-up activities to promote alternative methods and address knowledge gaps. During the development of its Recommendation, EURL ECVAM typically mandates the EURL ECVAM Scientific Advisory Committee (ESAC) to carry out an independent scientific peer review which is communicated as an ESAC Opinion and Working Group report. In addition, EURL ECVAM consults with other Commission services, EURL ECVAM’s advisory body for Preliminary Assessment of Regulatory Relevance (PARERE), the EURL ECVAM Stakeholder Forum (ESTAF) and with partner organisations of the International Collaboration on Alternative Test Methods (ICATM). Contact information European Commission, Joint Research Centre (JRC), Chemical Safety and Alternative Methods Unit (F3) Address: via E.
    [Show full text]
  • Monobodies and Other Synthetic Binding Proteins for Expanding Protein Science
    Reviews Protein Science DOI 10.1002/pro.3148 Monobodies and Other Synthetic Binding Proteins for Expanding Protein Science Fern Sha,1,4 Gabriel Salzman,1 Ankit Gupta1,2 and Shohei Koide1,2,3* 1Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA 2Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA 3Department of Biochemistry and Molecular Pharmacology New York University School of Medicine, New York, NY 10016, USA 4Present address, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20148, USA *Correspondence to: Shohei Koide, Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA. E-mail: [email protected] Abstract Synthetic binding proteins are constructed using non-antibody molecular scaffolds. Over the last two decades, in-depth structural and functional analyses of synthetic binding proteins have improved combinatorial library designs and selection strategies, which have resulted in potent platforms that consistently generate binding proteins to diverse targets with affinity and specificity that rival those of antibodies. Favorable attributes of synthetic binding proteins, such as small size, freedom from disulfide bond formation and ease of making fusion proteins, have enabled their unique applications in protein science, cell biology and beyond. Here, we review recent studies that illustrate how synthetic binding proteins are powerful probes that can directly link structure and function,
    [Show full text]
  • Highly Potent Anti-SARS-Cov-2 Multi-Darpin Therapeutic Candidates Marcel Walser1,*, Sylvia Rothenberger2,3,*, Daniel L
    bioRxiv preprint doi: https://doi.org/10.1101/2020.08.25.256339; this version posted August 26, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Highly potent anti-SARS-CoV-2 multi-DARPin therapeutic candidates Marcel Walser1,*, Sylvia Rothenberger2,3,*, Daniel L. Hurdiss4,5,*, Anja Schlegel1, Valérie Calabro1, Simon Fontaine1, Denis Villemagne1, Maria Paladino1, Tanja Hospodarsch1, Alexandra Neculcea1, Andreas Cornelius1, Patricia Schildknecht1, Mirela Matzner1, Martin Hänggi1, Marco Franchini1, Yvonne 5 Kaufmann1, Iris Schlegel1,, Chloe Iss1, Thamar Loser1, Susanne Mangold1, Christel Herzog1, Dieter Schiegg1, Christian Reichen1, Filip Radom1, Andreas Bosshart1, Andreas Lehmann1, Micha A. Haeuptle1, Alexander Zürcher1, Toni Vagt1, Gabriel Sigrist1, Marcel Straumann 1, Karl Proba1, Niina Veitonmäki1, Keith M. Dawson1, Christof Zitt1, Jennifer Mayor2,3, Sarah Ryter2, Heyrhyoung Lyoo4, Chunyan Wang4, Wentao Li4, Ieva Drulyte6, H. Kaspar Binz7, Leon de Waal8, Koert J. Stittelaar8, Seth Lewis1, Daniel 10 Steiner1, Frank J.M. van Kuppeveld4, Olivier Engler2, Berend-Jan Bosch4, Michael T. Stumpp1,9, Patrick Amstutz 1 *These authors contributed equally to this work 1Molecular Partners AG, Wagistrasse 14, 8952 Zurich-Schlieren, Switzerland 15 2Spiez Laboratory, Austrasse, 3700 Spiez, Switzerland 3Institute of Microbiology, University Hospital Center
    [Show full text]
  • (12) United States Patent (10) Patent No.: US 9,499,622 B2 Cheong Et Al
    USOO9499622B2 (12) United States Patent (10) Patent No.: US 9,499,622 B2 Cheong et al. (45) Date of Patent: Nov. 22, 2016 (54) ANTI-EGFR/ANTI-HER2 BISPECIFIC 5,821,337 A 10, 1998 Carter et al. ANTIBODIES WITH ANT-EGFR DARPNS 6,165,464 A 12/2000 Hudziak et al. 7,417,130 B2 8/2008 Stumpp et al. 8,071,730 B2 12/2011 Goetsch et al. (71) Applicant: Samsung Electronics Co., Ltd., 8, 110,653 B2 2/2012 Stumpp et al. Suwon-si, Gyeonggi-do (KR) 8,329,873 B2 12/2012 Adams et al. 8,524,244 B2 9/2013 Camphausen et al. (72) Inventors: Kwang Ho Cheong, Seoul (KR): 9,234,028 B2 1/2016 Camphausen et al. Seung Hyun Lee, Suwon-si (KR): 9,359.440 B2 * 6/2016 Cheong ................ CO7K 14,705 Powei Lin, Hwaseong-si (KR); Saet 2009/0010840 A1 1/2009 Adams et al. Byoul Lee, Seoul (KR); Jae Woong 2010/0178298 A1 7/2010 Lindhofer 2012fOO2O966 A1* 1/2012 Barbas, III ........... CO7K 14,515 Hwang, Seoul (KR) 424,134.1 2012/0156191 A1 6/2012 Goetsch et al. (73) Assignee: SAMSUNGELECTRONICS CO., 2012,0277143 A1 1 1/2012 Jacobs et al. LTD., Suwon-Si (KR) 2013, OO17200 A1 1/2013 Scheer et al. 2015.OO30596 A1* 1/2015 Cheong ................ CO7K 14,705 (*) Notice: Subject to any disclaimer, the term of this 424,134.1 patent is extended or adjusted under 35 U.S.C. 154(b) by 0 days. FOREIGN PATENT DOCUMENTS EP 1332.209 B2 11/2009 (21) Appl.
    [Show full text]
  • (10) Patent No.: US 9034822 B2
    USOO9034822B2 (12) United States Patent (10) Patent No.: US 9,034,822 B2 Van Ryn et al. (45) Date of Patent: *May 19, 2015 (54) METHODS OF USING ANTIBODIES DURING (56) References Cited ANTCOAGULANT THERAPY OF DABGATRAN AND/OR RELATED U.S. PATENT DOCUMENTS COMPOUNDS 6,440,417 B1 8/2002 Thibaudeau et al. 6,469,039 B1 10/2002 Hauel et al. (71) Applicants: Joanne Van Ryn, Warthausen (DE); 8,486,398 B2* 7/2013 Van Ryn et al. ........... 424,133.1 2004/OO97547 A1 5, 2004 Taveras et al. John Edward Park, Warthausen (DE): 2011/0206656 A1 8/2011 Van Ryn et al. Norbert Hauel, Schemmerhofen (DE): 2012fOO27780 A1 2/2012 Van Rynet al. Ulrich Kunz, Biberach an der Riss (DE); Tobias Litzenburger, Mittelbiberach FOREIGN PATENT DOCUMENTS (DE); Keith Canada, Southbury, CT CA 2 277 949 8, 1998 (US); Sanjaya Singh, Sandy Hook, CT CA 2277949 A1 * 8, 1998 (US); Alisa Waterman, Weston, CT WO WO-9837O75 8, 1998 (US) WO WO-2011 O23.653 3, 2011 WO WO-2011 089183 T 2011 (72) Inventors: Joanne Van Ryn, Warthausen (DE); John Edward Park, Warthausen (DE): OTHER PUBLICATIONS Norbert Hauel, Schemmerhofen (DE): Schlaeppi et al., Eur J Biochem. Mar. 10, 1990:188(2):463-70.* Ulrich Kunz, Biberach an der Riss (DE); Herion et al., Blood. May 1985:65(5):1201-7.* Tobias Litzenburger, Mittelbiberach Colburn, W. A., “Specific antibodies and fab fragments to alter the (DE); Keith Canada, Southbury, CT pharmacokinetics and reverse the pharmacologic/toxicologic effects (US); Sanjaya Singh, Sandy Hook, CT of drugs.” Drug Metabolism Reviews, 1980, vol.
    [Show full text]
  • WO 2017/147538 Al 31 August 2017 (31.08.2017) P O P C T
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization International Bureau (10) International Publication Number (43) International Publication Date WO 2017/147538 Al 31 August 2017 (31.08.2017) P O P C T (51) International Patent Classification: (81) Designated States (unless otherwise indicated, for every C12N 15/85 (2006.01) C12N 15/90 (2006.01) kind of national protection available): AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, (21) International Application Number: BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DJ, DK, DM, PCT/US2017/01953 1 DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, (22) International Filing Date: HN, HR, HU, ID, IL, IN, IR, IS, JP, KE, KG, KH, KN, 24 February 2017 (24.02.2017) KP, KR, KW, KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, (25) Filing Language: English NI, NO, NZ, OM, PA, PE, PG, PH, PL, PT, QA, RO, RS, (26) Publication Language: English RU, RW, SA, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, (30) Priority Data: ZA, ZM, ZW. 62/300,387 26 February 2016 (26.02.2016) U S (84) Designated States (unless otherwise indicated, for every (71) Applicant: POSEIDA THERAPEUTICS, INC. kind of regional protection available): ARIPO (BW, GH, [US/US]; 4242 Campus Point Ct #700, San Diego, Califor GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, nia 92121 (US).
    [Show full text]
  • Bi-Paratopic and Multivalent VH Domains Block ACE2 Binding and Neutralize SARS-Cov-2
    ARTICLES https://doi.org/10.1038/s41589-020-00679-1 Bi-paratopic and multivalent VH domains block ACE2 binding and neutralize SARS-CoV-2 Colton J. Bracken 1, Shion A. Lim 1, Paige Solomon1, Nicholas J. Rettko 1, Duy P. Nguyen1,5, Beth Shoshana Zha2, Kaitlin Schaefer1, James R. Byrnes 1, Jie Zhou1, Irene Lui 1, Jia Liu1,6, Katarina Pance1, QCRG Structural Biology Consortium*, Xin X. Zhou1, Kevin K. Leung 1 and James A. Wells 1,3,4 ✉ Neutralizing agents against SARS-CoV-2 are urgently needed for the treatment and prophylaxis of COVID-19. Here, we present a strategy to rapidly identify and assemble synthetic human variable heavy (VH) domains toward neutralizing epit- opes. We constructed a VH-phage library and targeted the angiotensin-converting enzyme 2 (ACE2) binding interface of the SARS-CoV-2 Spike receptor-binding domain (Spike-RBD). Using a masked selection approach, we identified VH binders to two non-overlapping epitopes and further assembled these into multivalent and bi-paratopic formats. These VH constructs showed increased affinity to Spike (up to 600-fold) and neutralization potency (up to 1,400-fold) on pseudotyped SARS-CoV-2 virus when compared to standalone VH domains. The most potent binder, a trivalent VH, neutralized authentic SARS-CoV-2 with a −1 half-maximal inhibitory concentration (IC50) of 4.0 nM (180 ng ml ). A cryo-EM structure of the trivalent VH bound to Spike shows each VH domain engaging an RBD at the ACE2 binding site, confirming our original design strategy. he emergence of SARS-CoV-2 and the associated COVID- Although this can be overcome with synthetic nanobody librar- 19 disease has emphasized the need to rapidly generate ies15,16, nanobody scaffolds that are animal-derived raise significant Ttherapeutics against novel pathogens.
    [Show full text]
  • Non-Invasive in Vivo Imaging of Tumour-Associated Cathepsin B by a Highly Selective Inhibitory Darpin
    SUPPLEMENTARY INFORMATION Non-invasive in vivo imaging of tumour-associated cathepsin B by a highly selective inhibitory DARPin Lovro Kramer1,2, Miha Renko1, Janja Završnik1,2, Dušan Turk1,3, Markus A. Seeger4, Olga Vasiljeva1, Markus G. Grütter5, Vito Turk1, Boris Turk1,3* 1 Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Ljubljana (Slovenia) 2 International Postgraduate School Jozef Stefan, Ljubljana (Slovenia) 3 Center of Excellence CIPKEBIP, Ljubljana (Slovenia) 4 Institute of Medical Microbiology, University of Zürich, Zürich (Switzerland) 5 Department of Biochemistry, University of Zürich, Zürich (Switzerland) *correspondence: [email protected] 1 SUPPLEMENTARY METHODS Expression and purification of recombinant cathepsins Human and mouse cathepsin B (CatB) were expressed in inclusion bodies of Escherichia coli and purified as described previously in [1] and [2], respectively. Other cathepsins were expressed in Pichia pastoris and purified as described previously for cathepsins L and S [3], cathepsin K [4] and cathepsin X [5]. Biotinylation of human CatB, mouse CatB and human CatS Recombinant human CatB, mouse CatB and human CatS were biotinylated with EZ-Link™ Sulfo-NHS-LC-LC-Biotin (Thermo Fisher Scientific) in PBS (pH 7.0) at 4°C for 2 h. The residual reagent was removed by gel filtration on PD10 columns (GE Healthcare Life Sciences), and the biotinylation efficiency was confirmed by Western blot and mass spectrometry (Fig. S16). Ribosome display selection of cathepsin B-specific inhibitory DARPins The aim of the study was to obtain DARPins that bind both human and mouse cathepsin B with high selectivity and affinity. We performed ribosome display of the recently described DARPin 2.0 library [6] against both CatB orthologues.
    [Show full text]
  • Type of the Paper (Article, Review, Communication
    Review Volume 11, Issue 3, 2021, 10679 - 10689 https://doi.org/10.33263/BRIAC113.1067910689 Selective Preference of Antibody Mimetics over Antibody, as Binding Molecules, for Diagnostic and Therapeutic Applications in Cancer Therapy Pankaj Garg 1,* 1 Department of Chemistry, GLA University, Mathura, 281406, India * Correspondence: [email protected]; Scopus Author ID 571962558738 Received: 5.10.2020; Revised: 3.11.2020; Accepted: 4.11.2020; Published: 7.11.2020 Abstract: Despite wider use of monoclonal and polyclonal antibodies as therapeutic and diagnostic detection agents for different types of cancers, their limitations for biomedical applications have forced scientists to design alternate next-generation molecular binding reagents, the so-called antibody mimetics. The ultimate aim to produce antibody mimetics is to out-perform the intrinsic limitations of antibodies related to their binding affinities, tumor penetration, temperature, and pH stability. The current review highlights the advanced characteristics and constructional modification of alternate antibody mimetics, compared to animal source generated antibodies and their improved applications in bioanalytical chemistry; especially in cancer treatment as a diagnostic and therapeutic tool. Keywords:Antibody mimetic; Monoclonal antibodies (MoAbs); Protein scaffold engineering; Molecular Imaging; cancer therapy. © 2020 by the authors. This article is an open-access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/). 1. Introduction Antibodies, especially monoclonal antibodies, on account of their high stability and specific affinity, have been identified as effective tools both for therapeutic and diagnostic applications, especially in cancer therapy. Antibodies are Y-shaped glycoproteins produced by the immune system to counteract the effect of any foreign substance or antigen in the body.
    [Show full text]
  • Highly Potent Anti-SARS-Cov-2 Multi-Darpin Therapeutic Candidates Marcel Walser1,*, Sylvia Rothenberger2,3,*, Daniel L
    bioRxiv preprint doi: https://doi.org/10.1101/2020.08.25.256339; this version posted November 20, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Highly potent anti-SARS-CoV-2 multi-DARPin therapeutic candidates Marcel Walser1,*, Sylvia Rothenberger2,3,*, Daniel L. Hurdiss4,5,*, Anja Schlegel1, Valérie Calabro1, Simon Fontaine1, Denis Villemagne1, Maria Paladino1, Tanja Hospodarsch1, Alexandra Neculcea1, Andreas Cornelius1, Patricia Schildknecht1, Mirela Matzner1, Martin Hänggi1, Marco Franchini1, Yvonne 5 Kaufmann1, Doris Schaible1, Iris Schlegel1, Chloe Iss1, Thamar Looser1, Susanne Mangold1, Christel Herzog1, Dieter Schiegg1, Christian Reichen1, Filip Radom1, Andreas Bosshart1, Andreas Lehmann1, Micha A. Haeuptle1, Alexander Zürcher1, Toni Vagt1, Gabriel Sigrist1, Marcel Straumann 1, Karl Proba1, Niina Veitonmäki1, Keith M. Dawson1, Christof Zitt1, Jennifer Mayor2,3, Sarah Ryter2, Heyrhyoung Lyoo4, Chunyan Wang4, Wentao Li4, Ieva Drulyte6, Wenjuan Du4, H. Kaspar Binz7, Leon de Waal8, Koert J. 10 Stittelaar8*, Sarah Taplin9, Seth Lewis1, Daniel Steiner1, Frank J.M. van Kuppeveld4, Olivier Engler2, Berend-Jan Bosch4, Michael T. Stumpp1,10, Patrick Amstutz 1 *These authors contributed equally to this work 1Molecular Partners AG, Wagistrasse 14, 8952 Zurich-Schlieren, Switzerland 15 2Spiez Laboratory, Austrasse, 3700 Spiez, Switzerland 3Institute
    [Show full text]