57 results on '"Aaron K. Sato"'
Search Results
2. Bivalent intra-spike binding provides durability against emergent Omicron lineages: Results from a global consortium
- Author
-
Heather M. Callaway, Kathryn M. Hastie, Sharon L. Schendel, Haoyang Li, Xiaoying Yu, Jeremy Shek, Tierra Buck, Sean Hui, Dan Bedinger, Camille Troup, S. Moses Dennison, Kan Li, Michael D. Alpert, Charles C. Bailey, Sharon Benzeno, Jody L. Bonnevier, Jin-Qiu Chen, Charm Chen, Hyeseon Cho, Peter D. Crompton, Vincent Dussupt, Kevin C. Entzminger, Yassine Ezzyat, Jonathan K. Fleming, Nick Geukens, Amy E. Gilbert, Yongjun Guan, Xiaojian Han, Christopher J. Harvey, Julia M. Hatler, Bryan Howie, Chao Hu, Ailong Huang, Maya Imbrechts, Aishun Jin, Nik Kamachi, Gladys Keitany, Mark Klinger, Jay K. Kolls, Shelly J. Krebs, Tingting Li, Feiyan Luo, Toshiaki Maruyama, Michael A. Meehl, Letzibeth Mendez-Rivera, Andrea Musa, C.J. Okumura, Benjamin E.R. Rubin, Aaron K. Sato, Meiying Shen, Anirudh Singh, Shuyi Song, Joshua Tan, Jeffrey M. Trimarchi, Dhruvkumar P. Upadhyay, Yingming Wang, Lei Yu, Tom Z. Yuan, Erik Yusko, Bjoern Peters, Georgia Tomaras, and Erica Ollmann Saphire
- Subjects
CP: Immunology ,Biology (General) ,QH301-705.5 - Abstract
Summary: The SARS-CoV-2 Omicron variant of concern (VoC) and its sublineages contain 31–36 mutations in spike and escape neutralization by most therapeutic antibodies. In a pseudovirus neutralization assay, 66 of the nearly 400 candidate therapeutics in the Coronavirus Immunotherapeutic Consortium (CoVIC) panel neutralize Omicron and multiple Omicron sublineages. Among natural immunoglobulin Gs (IgGs), especially those in the receptor-binding domain (RBD)-2 epitope community, nearly all Omicron neutralizers recognize spike bivalently, with both antigen-binding fragments (Fabs) simultaneously engaging adjacent RBDs on the same spike. Most IgGs that do not neutralize Omicron bind either entirely monovalently or have some (22%–50%) monovalent occupancy. Cleavage of bivalent-binding IgGs to Fabs abolishes neutralization and binding affinity, with disproportionate loss of activity against Omicron pseudovirus and spike. These results suggest that VoC-resistant antibodies overcome mutagenic substitution via avidity. Hence, vaccine strategies targeting future SARS-CoV-2 variants should consider epitope display with spacing and organization identical to trimeric spike.
- Published
- 2023
- Full Text
- View/download PDF
3. Rapid discovery of diverse neutralizing SARS-CoV-2 antibodies from large-scale synthetic phage libraries
- Author
-
Tom Z. Yuan, Pankaj Garg, Linya Wang, Jordan R. Willis, Eric Kwan, Ana G Lujan Hernandez, Emily Tuscano, Emily N. Sever, Erica Keane, Cinque Soto, Eric M. Mucker, Mallorie E. Fouch, Edgar Davidson, Benjamin J. Doranz, Shweta Kailasan, M. Javad Aman, Haoyang Li, Jay W. Hooper, Erica Ollmann Saphire, James E. Crowe, Qiang Liu, Fumiko Axelrod, and Aaron K. Sato
- Subjects
sars-cov-2 ,neutralizing antibody ,covid-19 ,spike glycoprotein ,synthetic libraries ,Therapeutics. Pharmacology ,RM1-950 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Coronavirus disease 2019 (COVID-19) is an evolving global public health crisis in need of therapeutic options. Passive immunization of monoclonal antibodies (mAbs) represents a promising therapeutic strategy capable of conferring immediate protection from SARS-CoV-2 infection. Herein, we describe the discovery and characterization of neutralizing SARS-CoV-2 IgG and VHH antibodies from four large-scale phage libraries. Each library was constructed synthetically with shuffled complementarity-determining region loops from natural llama and human antibody repertoires. While most candidates targeted the receptor-binding domain of the S1 subunit of SARS-CoV-2 spike protein, we also identified a neutralizing IgG candidate that binds a unique epitope on the N-terminal domain. A select number of antibodies retained binding to SARS-CoV-2 variants Alpha, Beta, Gamma, Kappa and Delta. Overall, our data show that synthetic phage libraries can rapidly yield SARS-CoV-2 S1 antibodies with therapeutically desirable features, including high affinity, unique binding sites, and potent neutralizing activity in vitro, and a capacity to limit disease in vivo.
- Published
- 2022
- Full Text
- View/download PDF
4. Functional GLP-1R antibodies identified from a synthetic GPCR-focused library demonstrate potent blood glucose control
- Author
-
Qiang Liu, Pankaj Garg, Burcu Hasdemir, Linya Wang, Emily Tuscano, Emily Sever, Erica Keane, Ana G Lujan Hernandez, Tom Z. Yuan, Eric Kwan, Joyce Lai, Greg Szot, Sreenivasan Paruthiyil, Fumiko Axelrod, and Aaron K. Sato
- Subjects
GPCR ,glp-1 peptide ,phage display library ,panning ,antagonist ,agonist ,Therapeutics. Pharmacology ,RM1-950 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
G protein-coupled receptors (GPCRs) are a group of seven-transmembrane receptor proteins that have proven to be successful drug targets. Antibodies are becoming an increasingly promising modality to target these receptors due to their unique properties, such as exquisite specificity, long half-life, and fewer side effects, and their improved pharmacokinetic and pharmacodynamic profiles compared to peptides and small molecules, which results from their more favorable biodistribution. To date, there are only two US Food and Drug Administration-approved GPCR antibody drugs, namely erenumab and mogamulizumab, and this highlights the challenges encountered in identifying functional antibodies against GPCRs. Utilizing Twist’s precision DNA writing technologies, we have created a GPCR-focused phage display library with 1 × 1010 diversity. Specifically, we mined endogenous GPCR binding ligand and peptide sequences and incorporated these binding motifs into the heavy chain complementarity-determining region 3 in a synthetic antibody library. Glucagon-like peptide-1 receptor (GLP-1 R) is a class B GPCR that acts as the receptor for the incretin GLP-1, which is released to regulate insulin levels in response to food intake. GLP-1 R agonists have been widely used to increase insulin secretion to lower blood glucose levels for the treatment of type 1 and type 2 diabetes, whereas GLP-1 R antagonists have applications in the treatment of severe hypoglycemia associated with bariatric surgery and hyperinsulinomic hypoglycemia. Here we present the discovery and creation of both antagonistic and agonistic GLP-1 R antibodies by panning this GPCR-focused phage display library on a GLP-1 R-overexpressing Chinese hamster ovary cell line and demonstrate their in vitro and in vivo functional activity.
- Published
- 2021
- Full Text
- View/download PDF
5. RF1 attenuation enables efficient non-natural amino acid incorporation for production of homogeneous antibody drug conjugates
- Author
-
Gang Yin, Heather T. Stephenson, Junhao Yang, Xiaofan Li, Stephanie M. Armstrong, Tyler H. Heibeck, Cuong Tran, Mary Rose Masikat, Sihong Zhou, Ryan L. Stafford, Alice Y. Yam, John Lee, Alexander R. Steiner, Avinash Gill, Kalyani Penta, Sonia Pollitt, Ramesh Baliga, Christopher J. Murray, Christopher D. Thanos, Leslie M. McEvoy, Aaron K. Sato, and Trevor J. Hallam
- Subjects
Medicine ,Science - Abstract
Abstract Amber codon suppression for the insertion of non-natural amino acids (nnAAs) is limited by competition with release factor 1 (RF1). Here we describe the genome engineering of a RF1 mutant strain that enhances suppression efficiency during cell-free protein synthesis, without significantly impacting cell growth during biomass production. Specifically, an out membrane protease (OmpT) cleavage site was engineered into the switch loop of RF1, which enables its conditional inactivation during cell lysis. This facilitates extract production without additional processing steps, resulting in a scaleable extract production process. The RF1 mutant extract allows nnAA incorporation at previously intractable sites of an IgG1 and at multiple sites in the same polypeptide chain. Conjugation of cytotoxic agents to these nnAAs, yields homogeneous antibody drug conjugates (ADCs) that can be optimized for conjugation site, drug to antibody ratio (DAR) and linker-warheads designed for efficient tumor killing. This platform provides the means to generate therapeutic ADCs inaccessible by other methods that are efficient in their cytotoxin delivery to tumor with reduced dose-limiting toxicities and thus have the potential for better clinical impact.
- Published
- 2017
- Full Text
- View/download PDF
6. Post-Exposure Protection in Mice against Sudan Virus by a Two Antibody Cocktail
- Author
-
Jeffrey W. Froude, Andrew S. Herbert, Thibaut Pelat, Sebastian Miethe, Samantha E. Zak, Jennifer M. Brannan, Russell R. Bakken, Alexander R. Steiner, Gang Yin, Trevor J. Hallam, Aaron K. Sato, Michael Hust, Philippe Thullier, and John M. Dye
- Subjects
Sudan virus ,Ebola ,antibody ,protection ,biodefense ,cell-free production ,phage display ,Microbiology ,QR1-502 - Abstract
Sudan virus (SUDV) and Ebola viruses (EBOV) are both members of the Ebolavirus genus and have been sources of epidemics and outbreaks for several decades. We present here the generation and characterization of cross-reactive antibodies to both SUDV and EBOV, which were produced in a cell-free system and protective against SUDV in mice. A non-human primate, cynomolgus macaque, was immunized with viral-replicon particles expressing the glycoprotein of SUDV-Boniface (8A). Two separate antibody fragment phage display libraries were constructed after four immunogen injections. Both libraries were screened first against the SUDV and a second library was cross-selected against EBOV-Kikwit. Sequencing of 288 selected clones from the two distinct libraries identified 58 clones with distinct VH and VL sequences. Many of these clones were cross-reactive to EBOV and SUDV and able to neutralize SUDV. Three of these recombinant antibodies (X10B1, X10F3, and X10H2) were produced in the scFv-Fc format utilizing a cell-free production system. Mice that were challenged with SUDV-Boniface receiving 100µg of the X10B1/X10H2 scFv-Fc combination 6 and 48-h post-exposure demonstrated partial protection individually and complete protection as a combination. The data herein suggests these antibodies may be promising candidates for further therapeutic development.
- Published
- 2018
- Full Text
- View/download PDF
7. Discovery of Fully Human Anti-MET Monoclonal Antibodies with Antitumor Activity against Colon Cancer Tumor Models In Vivo
- Author
-
Edward Htun van der Horst, Lawrence Chinn, Min Wang, Timothy Velilla, Hoang Tran, Yarrow Madrona, Andrew Lam, May Ji, Timothy C. Hoey, and Aaron K. Sato
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The receptor tyrosine kinase MET is a major component controlling the invasive growth program in embryonic development and in invasive malignancies. The discovery of therapeutic antibodies against MET has been difficult, and antibodies that compete with hepatocyte growth factor (HGF) act as agonists. By applying phage technology and cell-based panning strategies, we discovered two fully human antibodies against MET (R13 and R28), which synergistically inhibit HGF binding to MET and elicit antibody-dependent cellular cytotoxicity. Cell-based phosphorylation assays demonstrate that R13 and R28 abrogate HGF-induced activation of MET, AKT1, ERK1/2, and HGF-induced migration and proliferation. FACS experiments suggest that the inhibitory effect is mediated by “locking” MET receptor in a state with R13, which then increases avidity of R28 for the extracellular domain of MET, thus blocking HGF binding without activating the receptor. In vivo studies demonstrate that the combination of R13/28 significantly inhibited tumor growth in various colon tumor xenograft models. Inhibition of tumor growth was associated with induction of hypoxia. Global gene expression analysis shows that inhibition of HGF/MET pathway significantly upregulated the tumor suppressors KLF6, CEACAM1, and BMP2, the negative regulator of phosphatidylinositol-3-OH-kinase PIK3IP1, and significantly suppressed SCF and SERPINE2, both enhancers of proliferation and invasiveness. Moreover, in an experimental metastasis model, R13/28 increased survival by preventing the recurrence of otherwise lethal lung metastases. Taken together, these results underscore the utility of a dual-antibody approach for targeting MET and possibly other receptor tyrosine kinases. Our approach could be expanded to drug discovery efforts against other cell surface proteins.
- Published
- 2009
- Full Text
- View/download PDF
8. Discovery and design of G protein-coupled receptor targeting antibodies
- Author
-
Sean M. Peterson, Catherine J. Hutchings, Cameron F. Hu, Melina Mathur, Janelle W. Salameh, Fumiko Axelrod, and Aaron K. Sato
- Subjects
Drug Discovery - Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Data from Discovery of STRO-002, a Novel Homogeneous ADC Targeting Folate Receptor Alpha, for the Treatment of Ovarian and Endometrial Cancers
- Author
-
Trevor J. Hallam, Kristin Bedard, Mark Lupher, Aaron K. Sato, Venita I. De Almeida, Toni Kline, Alice Y. Yam, Ryan L. Stafford, Gang Yin, Cuong Tran, Daniel Calarese, Tyler H. Heibeck, Jeffrey Hanson, Robert Henningsen, Heather T. Stephenson, Krishna Bajjuri, Jennifer Smith, Stellanie Krimm, Cristina L. Abrahams, Sihong Zhou, and Xiaofan Li
- Abstract
supplementary text and figures
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Discovery of STRO-002, a Novel Homogeneous ADC Targeting Folate Receptor Alpha, for the Treatment of Ovarian and Endometrial Cancers
- Author
-
Trevor J. Hallam, Kristin Bedard, Mark Lupher, Aaron K. Sato, Venita I. De Almeida, Toni Kline, Alice Y. Yam, Ryan L. Stafford, Gang Yin, Cuong Tran, Daniel Calarese, Tyler H. Heibeck, Jeffrey Hanson, Robert Henningsen, Heather T. Stephenson, Krishna Bajjuri, Jennifer Smith, Stellanie Krimm, Cristina L. Abrahams, Sihong Zhou, and Xiaofan Li
- Abstract
STRO-002 is a novel homogeneous folate receptor alpha (FolRα) targeting antibody–drug conjugate (ADC) currently being investigated in the clinic as a treatment for ovarian and endometrial cancers. Here, we describe the discovery, optimization, and antitumor properties of STRO-002. STRO-002 was generated by conjugation of a novel cleavable 3-aminophenyl hemiasterlin linker-warhead (SC239) to the nonnatural amino acid para-azidomethyl-L-phenylalanine incorporated at specific positions within a high affinity anti-FolRα antibody using Sutro's XpressCF+, which resulted in a homogeneous ADC with a drug–antibody ratio (DAR) of 4. STRO-002 binds to FolRα with high affinity, internalizes rapidly into target positive cells, and releases the tubulin-targeting cytotoxin 3-aminophenyl hemiasterlin (SC209). SC209 has reduced potential for drug efflux via P-glycoprotein 1 drug pump compared with other tubulin-targeting payloads. While STRO-002 lacks nonspecific cytotoxicity toward FolRα-negative cell lines, bystander killing of target negative cells was observed when cocultured with target positive cells. STRO-002 is stable in circulation with no change in DAR for up to 21 days and has a half-life of 6.4 days in mice. A single dose of STRO-002 induced significant tumor growth inhibition in FolRα-expressing xenograft models and patient-derived xenograft models. In addition, combination treatment with carboplatin or Avastin further increased STRO-002 efficacy in xenograft models. The potent and specific preclinical efficacy of STRO-002 supports clinical development of STRO-002 for treating patients with FolRα-expressing cancers, including ovarian, endometrial, and non–small cell lung cancer. Phase I dose escalation for STRO-002 is in progress in ovarian cancer and endometrial cancer patients (NCT03748186 and NCT05200364).
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Information: Supplementary Figures 1-5 from Targeted Drug Delivery with an Integrin-Binding Knottin–Fc–MMAF Conjugate Produced by Cell-Free Protein Synthesis
- Author
-
Jennifer R. Cochran, Aaron K. Sato, Alexander Steiner, Maria Filsinger Interrante, Rishard Chen, James R. Kintzing, Shelley E. Ackerman, and Nicolas V. Currier
- Abstract
Supplementary Figure 1. Standard curves for quantification of cellular integrin expression; Supplementary Figure 2. Competition binding to U87MG tumor cell surface integrins; Supplementary Figure 3. Inhibition of tumor cell proliferation. U87MG, A2780 and MB-468 cells were incubated with varying concentrations of 2.5F-Fc-MMAF, and CTRL-Fc-MMAF;Supplementary Figure 4. Growth inhibition of subcutaneous U87MG tumors implanted into Nu/Nu mice; Supplementary Figure 5. Weight monitoring.
- Published
- 2023
- Full Text
- View/download PDF
12. A General Sequence Processing and Analysis Program for Protein Engineering.
- Author
-
Ryan L. Stafford, Erik S. Zimmerman, Trevor J. Hallam, and Aaron K. Sato
- Published
- 2014
- Full Text
- View/download PDF
13. A synthetic bispecific antibody capable of neutralizing SARS-CoV-2 Delta and Omicron
- Author
-
Tom Z. Yuan, Carolina Lucas, Valter S. Monteiro, Akiko Iwasaki, Marisa L. Yang, Hector F. Nepita, Ana G. Lujan Hernandez, Joseph M. Taft, Lester Frei, Sai T. Reddy, Cédric R. Weber, Kevin P. Malobisky, Rodrigo Mesquita, and Aaron K. Sato
- Subjects
virus diseases - Abstract
Bispecific antibodies have emerged as a promising strategy for curtailing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immune escape. This brief report highlights RBT-0813 (also known as TB493-04), a synthetic, humanized, receptor-binding domain (RBD)-targeted bispecific antibody that retains picomolar affinity to the Spike (S) trimers of all major variants of concern and neutralizes both SARS-CoV-2 Delta and Omicron in vitro.
- Published
- 2022
- Full Text
- View/download PDF
14. Rapid discovery of diverse neutralizing SARS-CoV-2 antibodies from large-scale synthetic phage libraries
- Author
-
Tom Z. Yuan, Pankaj Garg, Linya Wang, Jordan R. Willis, Eric Kwan, Ana G Lujan Hernandez, Emily Tuscano, Emily N. Sever, Erica Keane, Cinque Soto, Eric M. Mucker, Mallorie E. Fouch, Edgar Davidson, Benjamin J. Doranz, Shweta Kailasan, M. Javad Aman, Haoyang Li, Jay W. Hooper, Erica Ollmann Saphire, James E. Crowe, Qiang Liu, Fumiko Axelrod, and Aaron K. Sato
- Subjects
viruses ,Immunology ,RM1-950 ,Antibodies, Viral ,Epitopes ,Antibody Specificity ,Peptide Library ,Report ,Chlorocebus aethiops ,Immunology and Allergy ,Animals ,Vero Cells ,spike glycoprotein ,Mesocricetus ,SARS-CoV-2 ,neutralizing antibody ,COVID-19 ,RC581-607 ,Single-Domain Antibodies ,Antibodies, Neutralizing ,synthetic libraries ,Disease Models, Animal ,Immunoglobulin G ,Host-Pathogen Interactions ,Spike Glycoprotein, Coronavirus ,Female ,Therapeutics. Pharmacology ,Binding Sites, Antibody ,Immunologic diseases. Allergy ,Cell Surface Display Techniques - Abstract
Coronavirus disease 2019 (COVID-19) is an evolving global public health crisis in need of therapeutic options. Passive immunization of monoclonal antibodies (mAbs) represents a promising therapeutic strategy capable of conferring immediate protection from SARS-CoV-2 infection. Herein, we describe the discovery and characterization of neutralizing SARS-CoV-2 IgG and VHH antibodies from four large-scale phage libraries. Each library was constructed synthetically with shuffled complementarity-determining region loops from natural llama and human antibody repertoires. While most candidates targeted the receptor-binding domain of the S1 subunit of SARS-CoV-2 spike protein, we also identified a neutralizing IgG candidate that binds a unique epitope on the N-terminal domain. A select number of antibodies retained binding to SARS-CoV-2 variants Alpha, Beta, Gamma, Kappa and Delta. Overall, our data show that synthetic phage libraries can rapidly yield SARS-CoV-2 S1 antibodies with therapeutically desirable features, including high affinity, unique binding sites, and potent neutralizing activity in vitro, and a capacity to limit disease in vivo.
- Published
- 2021
15. Defining variant-resistant epitopes targeted by SARS-CoV-2 antibodies: A global consortium study
- Author
-
Yanan Lu, Timothy C. Germann, Adrian Enriquez, Ruben Diaz Avalos, Marit J. van Gils, Ana G. Lujan Hernandez, Yoann Aldon, Guojun Lang, Mohammad M. Sajadi, Elizabeth Feeney, John Ingraham, Georgia D. Tomaras, Milite Abraha, Junli Liu, Jacob Glanville, S. Moses Dennison, Minsoo Kim, Erica Ollmann Saphire, Sawsan Youssef, Ross S. Federman, Gillian Q. Horn, Bingqing Shen, Sean Hui, Anne L. Palser, Joseph M. Fernandez, Gevorg Grigoryan, Vamseedhar Rayaprolu, David D. Ho, Jieyun Yin, Sharon L. Schendel, Jian Yu, Haoyang Li, Shelly J. Krebs, Qian Zhang, Ronald R. Cobb, Colin Mann, Eduardo Olmedillas, Rogier W. Sanders, Robin Green, Hyeong Mi Kim, Tierra Buck, Dawid Zyla, Hanif Ali, Paul Kellam, Terence H. Rabbitts, Diptiben V. Parekh, Anna J. MacCamy, Cheolmin Kim, Kuan-Ying A. Huang, Milan T. Tomic, Andrew T. McGuire, Tom Z. Yuan, Chitra Hariharan, Zahra Rikhtegaran Tehrani, Bjoern Peters, Xiaoying Yu, Sooyoung Lee, CoVIC-DB team, Kan Li, Leonidas Stamatatos, Cheuk Lun Leung, Chao Kong, Michelle Zandonatti, Fei Lan, Yongcong Tan, Aaron K. Sato, Kathryn M. Hastie, Liang Schweizer, Sanja Aracic, Jonne L. Snitselaar, Weidong Jiang, Kelly Shaffer, Jimin Seo, Daniel Bedinger, Medical Microbiology and Infection Prevention, and AII - Infectious diseases
- Subjects
2019-20 coronavirus outbreak ,Multidisciplinary ,Coronavirus disease 2019 (COVID-19) ,biology ,Immunodominant Epitopes ,SARS-CoV-2 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Protein domain ,COVID-19 ,Antibodies, Viral ,Virology ,Antibodies, Neutralizing ,Epitope ,Article ,Epitope mapping ,Protein Domains ,Spike Glycoprotein, Coronavirus ,biology.protein ,Humans ,Antibody ,Antigens, Viral ,Epitope Mapping ,Protein Binding - Abstract
Antibody-based therapeutics and vaccines are essential to combat COVID-19 morbidity and mortality after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Multiple mutations in SARS-CoV-2 that could impair antibody defenses propagated in human-to-human transmission and spillover or spillback events between humans and animals. To develop prevention and therapeutic strategies, we formed an international consortium to map the epitope landscape on the SARS-CoV-2 spike protein, defining and structurally illustrating seven receptor binding domain (RBD)–directed antibody communities with distinct footprints and competition profiles. Pseudovirion-based neutralization assays reveal spike mutations, individually and clustered together in variants, that affect antibody function among the communities. Key classes of RBD-targeted antibodies maintain neutralization activity against these emerging SARS-CoV-2 variants. These results provide a framework for selecting antibody treatment cocktails and understanding how viral variants might affect antibody therapeutic efficacy.
- Published
- 2021
16. Rapid exploration of the epitope coverage produced by an Ebola survivor to guide the discovery of therapeutic antibody cocktails
- Author
-
Tom Z. Yuan, Mohammad Javad Aman, Fumiko Takada Axelrod, Erica Keane, Aaron K. Sato, Yasmina N Abdiche, Madeleine Noonan-Shueh, Qiang Liu, Shweta Kailasan, and Ana G Lujan Hernandez
- Subjects
0301 basic medicine ,infectious disease ,Immunology ,Computational biology ,Biology ,medicine.disease_cause ,Neutralization ,Epitope ,Ebola virus ,03 medical and health sciences ,Synthetic biology ,0302 clinical medicine ,Antigen ,Epitope binning ,medicine ,Immunology and Allergy ,neutralizing antibodies ,Original Research Article ,antibody therapeutics ,AcademicSubjects/SCI01030 ,030104 developmental biology ,Infectious disease (medical specialty) ,030220 oncology & carcinogenesis ,biology.protein ,AcademicSubjects/SCI00100 ,synthetic biology ,Antibody ,surface plasmon resonance - Abstract
Background Development of successful neutralizing antibodies is dependent upon broad epitope coverage to increase the likelihood of achieving therapeutic function. Recent advances in synthetic biology have allowed us to conduct an epitope binning study on a large panel of antibodies identified to bind to Ebola virus glycoprotein with only published sequences. Methods and Results A rapid, first-pass epitope binning experiment revealed seven distinct epitope families that overlapped with known structural epitopes from the literature. A focused set of antibodies was selected from representative clones per bin to guide a second-pass binning that revealed previously unassigned epitopes, confirmed epitopes known to be associated with neutralizing antibodies, and demonstrated asymmetric blocking of EBOV GP from allosteric effectors reported from literature. Conclusions Critically, this workflow allows us to probe the epitope landscape of EBOV GP without any prior structural knowledge of the antigen or structural benchmark clones. Incorporating epitope binning on hundreds of antibodies during early stage antibody characterization ensures access to a library’s full epitope coverage, aids in the identification of high quality reagents within the library that recapitulate this diversity for use in other studies, and ultimately enables the rational development of therapeutic cocktails that take advantage of multiple mechanisms of action such as cooperative synergistic effects to enhance neutralization function and minimize the risk of mutagenic escape. The use of high-throughput epitope binning during new outbreaks such as the current COVID-19 pandemic is particularly useful in accelerating timelines due to the large amount of information gained in a single experiment., Starting with only published antibody sequences and soluble Ebola virus glycoprotein, high-throughput surface plasmon resonance is employed to rapidly screen and epitope bin hundreds of antibodies to fully characterize the epitope coverage of antibodies identified from a convalescent donor.
- Published
- 2020
17. Malaria Derived Glycosylphosphatidylinositol Anchor Enhances Anti-Pfs25 Functional Antibodies That Block Malaria Transmission
- Author
-
Wei Chan, Ashley J. Birkett, Gang Yin, Alexander Steiner, Neeraj Kapoor, Cuong D. Tran, Aaron K. Sato, Ivana Vanjak, Aym Berges, Kazutoyo Miura, Jeff Fairman, Carole A. Long, Thao P. Pham, and James Rozzelle
- Subjects
0301 basic medicine ,Glycosylphosphatidylinositols ,030231 tropical medicine ,Plasmodium falciparum ,Protozoan Proteins ,Biology ,medicine.disease_cause ,Biochemistry ,law.invention ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,law ,Conjugate vaccine ,parasitic diseases ,Malaria Vaccines ,medicine ,Animals ,Humans ,Vector (molecular biology) ,Artemisinin ,Malaria, Falciparum ,Escherichia coli ,Vaccines, Synthetic ,Vaccines, Conjugate ,Communication ,medicine.disease ,Virology ,Malaria ,030104 developmental biology ,Antibody Formation ,Recombinant DNA ,biology.protein ,Immunization ,Antibody ,medicine.drug - Abstract
Malaria, one of the most common vector borne human diseases, is a major world health issue. In 2015 alone, more than 200 million people were infected with malaria, out of which, 429 000 died. Even though artemisinin-based combination therapies (ACT) are highly effective at treating malaria infections, novel efforts toward development of vaccines to prevent transmission are still needed. Pfs25, a postfertilization stage parasite surface antigen, is a leading transmission-blocking vaccine (TBV) candidate. It is postulated that Pfs25 anchors to the cell membrane using a glycosylphosphatidylinositol (GPI) linker, which itself possesses pro-inflammatory properties. In this study, Escherichia coli derived extract (XtractCF+TM) was used in cell free protein synthesis [CFPS] to successfully express >200 mg/L of recombinant Pfs25 with a C-terminal non-natural amino acid (nnAA), namely, p-azidomethyl phenylalanine (pAMF), which possesses a reactive azide group. Thereafter, a unique conjugate vaccine (CV), namely, Pfs25-GPI was generated with dibenzocyclooctyne (DBCO) derivatized glycan core of malaria GPI using a simple but highly efficient copper free click chemistry reaction. In mice immunized with Pfs25 or Pfs25-GPI, the Pfs25-GPI group showed significantly higher titers compared to the Pfs25 group. Moreover, only purified IgGs from Pfs25-GPI group were able to significantly block transmission of parasites to mosquitoes, as judged by a standard membrane feeding assay [SMFA]. To our knowledge, this is the first report of the generation of a CV using Pfs25 and malaria specific GPI where the GPI is shown to enhance the ability of Pfs25 to elicit transmission blocking antibodies.
- Published
- 2018
18. RF1 attenuation enables efficient non-natural amino acid incorporation for production of homogeneous antibody drug conjugates
- Author
-
Alexander Steiner, Gang Yin, John K. Lee, Ramesh Baliga, Christopher J. Murray, Avinash Gill, Cuong D. Tran, Alice Yam, Junhao Yang, Sihong Zhou, Trevor J. Hallam, Sonia Pollitt, Christopher D. Thanos, Xiaofan Li, Stephanie Armstrong, Tyler H. Heibeck, Kalyani Penta, Ryan Stafford, Leslie Mcevoy, Aaron K. Sato, Heather Stephenson, and Mary Rose Masikat
- Subjects
Models, Molecular ,0301 basic medicine ,Immunoconjugates ,Lysis ,Cell Survival ,Protein Conformation ,medicine.medical_treatment ,Science ,Mutant ,Biology ,Protein Engineering ,Mass Spectrometry ,Article ,Structure-Activity Relationship ,03 medical and health sciences ,Drug Stability ,Cell Line, Tumor ,medicine ,Protein biosynthesis ,Humans ,Amino Acids ,chemistry.chemical_classification ,Binding Sites ,Multidisciplinary ,Protease ,Protein engineering ,Trastuzumab ,OmpT ,Amino acid ,030104 developmental biology ,Biochemistry ,chemistry ,Immunoglobulin G ,Mutation ,Codon, Terminator ,Medicine ,Release factor ,Chromatography, Liquid ,Peptide Termination Factors ,Protein Binding - Abstract
Amber codon suppression for the insertion of non-natural amino acids (nnAAs) is limited by competition with release factor 1 (RF1). Here we describe the genome engineering of a RF1 mutant strain that enhances suppression efficiency during cell-free protein synthesis, without significantly impacting cell growth during biomass production. Specifically, an out membrane protease (OmpT) cleavage site was engineered into the switch loop of RF1, which enables its conditional inactivation during cell lysis. This facilitates extract production without additional processing steps, resulting in a scaleable extract production process. The RF1 mutant extract allows nnAA incorporation at previously intractable sites of an IgG1 and at multiple sites in the same polypeptide chain. Conjugation of cytotoxic agents to these nnAAs, yields homogeneous antibody drug conjugates (ADCs) that can be optimized for conjugation site, drug to antibody ratio (DAR) and linker-warheads designed for efficient tumor killing. This platform provides the means to generate therapeutic ADCs inaccessible by other methods that are efficient in their cytotoxin delivery to tumor with reduced dose-limiting toxicities and thus have the potential for better clinical impact.
- Published
- 2017
19. RP-HPLC DAR Characterization of Site-Specific Antibody Drug Conjugates Produced in a Cell-Free Expression System
- Author
-
Trevor J. Hallam, Yiren Xu, Mary Rose Masikat, Gang Yin, Aaron K. Sato, Tyler H. Heibeck, Alexander Steiner, Cuong Tran, Qi Cai, Guifeng Jiang, and Xiaofan Li
- Subjects
0301 basic medicine ,biology ,Chemistry ,medicine.drug_class ,010401 analytical chemistry ,Organic Chemistry ,Lysine ,Monoclonal antibody ,01 natural sciences ,Small molecule ,0104 chemical sciences ,body regions ,03 medical and health sciences ,030104 developmental biology ,Biochemistry ,Pharmacokinetics ,PEG ratio ,medicine ,biology.protein ,Physical and Theoretical Chemistry ,Antibody ,Cytotoxicity ,Conjugate - Abstract
Antibody drug conjugates (ADCs) harness the target specificity of a monoclonal antibody (mAb) and the high cytotoxicity of a small molecule, enabling improved delivery of a potent antitumor agent compared to traditional chemotherapy for cancer therapy. Only two ADCs have been marketed, both of which are produced via nonsite-specific conjugation of the cytotoxic drug to either interchain cysteine (Adcetris) or lysine (Kadcyla). A growing body of evidence suggests that site-specific ADCs, because of their payload homogeneity, will improve pharmacokinetics and have wider therapeutic windows when compared to heterogeneous ADCs. Previously, we have demonstrated the use of a cell free expression system (Xpress CF+) for rapid production of site-specific ADCs. Here we report the generation of a variety of ADCs via conjugation between a site-specific incorporated non-natural amino acid (nnAA), para-azidomethyl-l-phenylalanine (pAMF), and dibenzocyclooctyl-(polyethylene glycol)4 (DBCO-(PEG)4) linked payloads using ...
- Published
- 2016
- Full Text
- View/download PDF
20. Targeted Drug Delivery with an Integrin-Binding Knottin–Fc–MMAF Conjugate Produced by Cell-Free Protein Synthesis
- Author
-
James R. Kintzing, Jennifer R. Cochran, Shelley Erin Ackerman, Nicolas V. Currier, Rishard Chen, Aaron K. Sato, Maria V. Filsinger Interrante, and Alexander Steiner
- Subjects
0301 basic medicine ,Integrins ,Cancer Research ,Immunoconjugates ,Cell Survival ,Integrin ,Pharmacology ,Mice ,03 medical and health sciences ,Drug Delivery Systems ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,Cell Proliferation ,Integrin binding ,Cell-free protein synthesis ,Cell-Free System ,biology ,Chemistry ,Cystine-Knot Miniproteins ,Xenograft Model Antitumor Assays ,Fusion protein ,Immunoglobulin Fc Fragments ,030104 developmental biology ,Oncology ,Targeted drug delivery ,030220 oncology & carcinogenesis ,Drug delivery ,Cancer cell ,biology.protein ,Peptides ,Oligopeptides ,Protein Binding ,Conjugate - Abstract
Antibody–drug conjugates (ADC) have generated significant interest as targeted therapeutics for cancer treatment, demonstrating improved clinical efficacy and safety compared with systemic chemotherapy. To extend this concept to other tumor-targeting proteins, we conjugated the tubulin inhibitor monomethyl-auristatin-F (MMAF) to 2.5F–Fc, a fusion protein composed of a human Fc domain and a cystine knot (knottin) miniprotein engineered to bind with high affinity to tumor-associated integrin receptors. The broad expression of integrins (including αvβ3, αvβ5, and α5β1) on tumor cells and their vasculature makes 2.5F-Fc an attractive tumor-targeting protein for drug delivery. We show that 2.5F-Fc can be expressed by cell-free protein synthesis, during which a non-natural amino acid was introduced into the Fc domain and subsequently used for site-specific conjugation of MMAF through a noncleavable linker. The resulting knottin–Fc–drug conjugate (KFDC), termed 2.5F-Fc-MMAF, had approximately 2 drugs attached per KFDC. 2.5F–Fc–MMAF inhibited proliferation in human glioblastoma (U87MG), ovarian (A2780), and breast (MB-468) cancer cells to a greater extent than 2.5F–Fc or MMAF alone or added in combination. As a single agent, 2.5F–Fc–MMAF was effective at inducing regression and prolonged survival in U87MG tumor xenograft models when administered at 10 mg/kg two times per week. In comparison, tumors treated with 2.5F–Fc or MMAF were nonresponsive, and treatment with a nontargeted control, CTRL–Fc–MMAF, showed a modest but not significant therapeutic effect. These studies provide proof-of-concept for further development of KFDCs as alternatives to ADCs for tumor targeting and drug delivery applications. Mol Cancer Ther; 15(6); 1291–300. ©2016 AACR.
- Published
- 2016
- Full Text
- View/download PDF
21. Post-Exposure Protection in Mice against Sudan Virus by a Two Antibody Cocktail
- Author
-
Sebastian Miethe, Aaron K. Sato, Alexander Steiner, Michael Hust, Russell R. Bakken, Jennifer M. Brannan, Philippe Thullier, Andrew S. Herbert, Trevor J. Hallam, Samantha E. Zak, Jeffrey W. Froude, Gang Yin, Thibaut Pelat, and John M. Dye
- Subjects
0301 basic medicine ,Male ,Phage display ,Immunogen ,cell-free production ,lcsh:QR1-502 ,Biology ,Cross Reactions ,medicine.disease_cause ,Antibodies, Viral ,Virus ,lcsh:Microbiology ,Article ,Viral Matrix Proteins ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Virology ,antibody ,medicine ,Animals ,Vaccines, Virus-Like Particle ,Ebolavirus ,Mice, Knockout ,Viral matrix protein ,Membrane Glycoproteins ,biodefense ,Vaccination ,Hemorrhagic Fever, Ebola ,protection ,Antibodies, Neutralizing ,Sudan virus ,030104 developmental biology ,Infectious Diseases ,Ebola ,phage display ,030220 oncology & carcinogenesis ,biology.protein ,Macaca ,Female ,Antibody ,Cell Surface Display Techniques ,Post-Exposure Prophylaxis ,Single-Chain Antibodies - Abstract
Sudan virus (SUDV) and Ebola viruses (EBOV) are both members of the Ebolavirus genus and have been sources of epidemics and outbreaks for several decades. We present here the generation and characterization of cross-reactive antibodies to both SUDV and EBOV, which were produced in a cell-free system and protective against SUDV in mice. A non-human primate, cynomolgus macaque, was immunized with viral-replicon particles expressing the glycoprotein of SUDV-Boniface (8A). Two separate antibody fragment phage display libraries were constructed after four immunogen injections. Both libraries were screened first against the SUDV and a second library was cross-selected against EBOV-Kikwit. Sequencing of 288 selected clones from the two distinct libraries identified 58 clones with distinct VH and VL sequences. Many of these clones were cross-reactive to EBOV and SUDV and able to neutralize SUDV. Three of these recombinant antibodies (X10B1, X10F3, and X10H2) were produced in the scFv-Fc format utilizing a cell-free production system. Mice that were challenged with SUDV-Boniface receiving 100µg of the X10B1/X10H2 scFv-Fc combination 6 and 48-h post-exposure demonstrated partial protection individually and complete protection as a combination. The data herein suggests these antibodies may be promising candidates for further therapeutic development.
- Published
- 2018
22. Toward a Ferrous Iron-Cleavable Linker for Antibody-Drug Conjugates
- Author
-
Aaron K. Sato, Sihong Zhou, Adam R. Renslo, James A. Wells, Toni Kline, Benjamin Spangler, Jeffrey A. Hanson, and Xiaofan Li
- Subjects
0301 basic medicine ,Immunoconjugates ,Receptor, ErbB-2 ,media_common.quotation_subject ,Iron ,Pharmaceutical Science ,Antineoplastic Agents ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Drug Discovery ,Cytotoxic T cell ,Moiety ,Animals ,Antigens ,Internalization ,media_common ,Mammals ,biology ,Chemistry ,Antibodies, Monoclonal ,Trastuzumab ,body regions ,030104 developmental biology ,Targeted drug delivery ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Cancer research ,Molecular Medicine ,Antibody ,Linker ,Conjugate - Abstract
Antibody-drug conjugates (ADCs) are antigen-targeted therapeutics that employ antibodies to deliver potent, cytotoxic effectors to cells with potentially high specificity. While promising clinical results have been achieved, significant pitfalls remain including internalization of ADCs in nontargeted cells expressing target antigen, which can limit therapeutic windows. Novel ADC linkers that are cleaved selectively in cancer cells but not in normal cells could minimize collateral damage caused by ADC uptake in nontargeted tissues. Here, we describe a prototypical ADC linker based on an Fe(II)-reactive 1,2,4-trioxolane scaffold (TRX) that by itself has demonstrated tumor-selective activity in preclinical cancer models. We prepared TRX-linked ADCs by site-selective conjugation to two sites in trastuzumab and compared their activity in Her2 positive and negative cells to ADC controls based on established linker chemistry. Our results confirm that the TRX moiety efficiently releases its payload following ADC uptake, affording picomolar potencies in antigen-positive cells. We also identified a destabilizing interaction between these initial TRX linkers and nearby antibody residues and suggest an approach to improve upon these prototypical designs.
- Published
- 2018
23. A simplified and robust protocol for immunoglobulin expression in E scherichia coli cell‐free protein synthesis systems
- Author
-
Cuong Tran, Rishard Chen, Aaron K. Sato, Jeffrey A. Hanson, Trevor J. Hallam, Mary Rose Masikat, Alexander R. Steiner, Gang Yin, Qi Cai, and James Zawada
- Subjects
Protein Folding ,protein synthesis ,IgG ,Coenzyme A ,Leucovorin ,Gene Expression ,Glutamic Acid ,Process Sensing and Control ,Biology ,medicine.disease_cause ,Protein Engineering ,chemistry.chemical_compound ,immunoglobulin G ,Viral Proteins ,RNA, Transfer ,expression ,Pyruvic Acid ,medicine ,Protein biosynthesis ,Escherichia coli ,Polyamines ,Putrescine ,T7 RNA polymerase ,cell‐free ,Cell-free protein synthesis ,Reproducibility of Results ,Protein engineering ,DNA-Directed RNA Polymerases ,Trastuzumab ,NAD ,Biochemistry ,chemistry ,Yield (chemistry) ,Protein Biosynthesis ,NAD+ kinase ,Biotechnology ,medicine.drug - Abstract
Cell‐free protein synthesis (CFPS) systems allow for robust protein expression with easy manipulation of conditions to improve protein yield and folding. Recent technological developments have significantly increased the productivity and reduced the operating costs of CFPS systems, such that they can compete with conventional in vivo protein production platforms, while also offering new routes for the discovery and production of biotherapeutics. As cell‐free systems have evolved, productivity increases have commonly been obtained by addition of components to previously designed reaction mixtures without careful re‐examination of the essentiality of reagents from previous generations. Here we present a systematic sensitivity analysis of the components in a conventional Escherichia coli CFPS reaction mixture to evaluate their optimal concentrations for production of the immunoglobulin G trastuzumab. We identify eight changes to the system, which result in optimal expression of trastuzumab. We find that doubling the potassium glutamate concentration, while entirely eliminating pyruvate, coenzyme A, NAD, total tRNA, folinic acid, putrescine and ammonium glutamate, results in a highly productive cell‐free system with a 95% reduction in reagent costs (excluding cell‐extract, plasmid, and T7 RNA polymerase made in‐house). A larger panel of other proteins was also tested and all show equivalent or improved yields with our simplified system. Furthermore, we demonstrate that all of the reagents for CFPS can be combined in a single freeze‐thaw stable master mix to improve reliability and ease of use. These improvements are important for the application of the CFPS system in fields such as protein engineering, high‐throughput screening, and biotherapeutics. © 2015 American Institute of Chemical Engineers Biotechnol. Prog., 31:823–831, 2015
- Published
- 2015
24. Methods to Make Homogenous Antibody Drug Conjugates
- Author
-
Kalyani Penta, Alexander R. Steiner, Aaron K. Sato, Gang Yin, Trevor J. Hallam, and Toni Kline
- Subjects
Drug ,Immunoconjugates ,media_common.quotation_subject ,Pharmacology toxicology ,Pharmaceutical Science ,Small Molecule Libraries ,Antibodies monoclonal ,Animals ,Humans ,Technology, Pharmaceutical ,Pharmacology (medical) ,media_common ,Pharmacology ,biology ,Chemistry ,Organic Chemistry ,targeted delivery ,Antibodies, Monoclonal ,antibody drug conjugates (ADCs) ,protein engineering ,site-specific conjugation ,homogenous conjugates ,Small molecule ,body regions ,Homogeneous ,Drug Design ,Immunology ,Cancer research ,biology.protein ,Molecular Medicine ,Antibody ,Expert Review ,Biotechnology ,Conjugate - Abstract
Antibody drug conjugates (ADCs) have progressed from hypothesis to approved therapeutics in less than 30 years, and the technologies available to modify both the antibodies and the cytotoxic drugs are expanding rapidly. For reasons well reviewed previously, the field is trending strongly toward homogeneous, defined antibody conjugation. In this review we present the antibody and small molecule chemistries that are currently used and being explored to develop specific, homogenous ADCs.
- Published
- 2014
- Full Text
- View/download PDF
25. A General Sequence Processing and Analysis Program for Protein Engineering
- Author
-
Trevor J. Hallam, Ryan Stafford, Erik S. Zimmerman, and Aaron K. Sato
- Subjects
Computer science ,General Chemical Engineering ,Molecular Sequence Data ,Library and Information Sciences ,Protein Engineering ,computer.software_genre ,User-Computer Interface ,Software ,Humans ,Gene Library ,Graphical user interface ,Electronic Data Processing ,Internet ,Base Sequence ,business.industry ,Experimental data ,Sequence Analysis, DNA ,General Chemistry ,File format ,Computer Science Applications ,Visualization ,Identification (information) ,Scripting language ,Trimming ,Data mining ,business ,Sequence Alignment ,computer - Abstract
Protein engineering projects often amass numerous raw DNA sequences, but no readily available software combines sequence processing and activity correlation required for efficient lead identification. XLibraryDisplay is an open source program integrated into Microsoft Excel for Windows that automates batch sequence processing via a simple step-by-step, menu-driven graphical user interface. XLibraryDisplay accepts any DNA template which is used as a basis for trimming, filtering, translating, and aligning hundreds to thousands of sequences (raw, FASTA, or Phred PHD file formats). Key steps for library characterization through lead discovery are available including library composition analysis, filtering by experimental data, graphing and correlating to experimental data, alignment to structural data extracted from PDB files, and generation of PyMOL visualization scripts. Though larger data sets can be handled, the program is best suited for analyzing approximately 10 000 or fewer leads or naïve clones which have been characterized using Sanger sequencing and other experimental approaches. XLibraryDisplay can be downloaded for free from sourceforge.net/projects/xlibrarydisplay/ .
- Published
- 2014
- Full Text
- View/download PDF
26. In vitro Fab display: a cell-free system for IgG discovery
- Author
-
Juan Jose Fung, Ryan Stafford, Gang Yin, Chris Murray, Xiaofan Li, Marissa L. Matsumoto, Alex Steiner, Ramesh Baliga, Trevor J. Hallam, Heather Stephenson, Qi Cai, Christopher D. Thanos, Willie D. Wang, Mary Rose Masikat, Monica You, Avinash Gill, Aaron K. Sato, Shwu-Hwa Lin, and Kalyani Penta
- Subjects
Vascular Endothelial Growth Factor A ,Fab selections ,Enzyme-Linked Immunosorbent Assay ,Bioengineering ,Antibodies, Monoclonal, Humanized ,Immunoglobulin light chain ,Biochemistry ,Antibodies ,Immunoglobulin G ,Immunoglobulin Fab Fragments ,Carcinoembryonic antigen ,Peptide Library ,Humans ,antibody reformatting ,Peptide library ,Molecular Biology ,Cell-free protein synthesis ,Cell-Free System ,biology ,ribosome display ,Original Articles ,Trastuzumab ,Molecular biology ,cell-free protein synthesis ,Carcinoembryonic Antigen ,Ribosome display ,biology.protein ,Antibody ,Cell Surface Display Techniques ,Biotechnology - Abstract
Selection technologies such as ribosome display enable the rapid discovery of novel antibody fragments entirely in vitro. It has been assumed that the open nature of the cell-free reactions used in these technologies limits selections to single-chain protein fragments. We present a simple approach for the selection of multi-chain proteins, such as antibody Fab fragments, using ribosome display. Specifically, we show that a two-chain trastuzumab (Herceptin) Fab domain can be displayed in a format which tethers either the heavy or light chain to the ribosome while retaining functional antigen binding. Then, we constructed synthetic Fab HC and LC libraries and performed test selections against carcinoembryonic antigen (CEA) and vascular endothelial growth factor (VEGF). The Fab selection output was reformatted into full-length immunoglobulin Gs (IgGs) and directly expressed at high levels in an optimized cell-free system for immediate screening, purification and characterization. Several novel IgGs were identified using this cell-free platform that bind to purified CEA, CEA positive cells and VEGF.
- Published
- 2014
- Full Text
- View/download PDF
27. Engineering toward a bacterial 'endoplasmic reticulum' for the rapid expression of immunoglobulin proteins
- Author
-
Evan Green, Ramesh Baliga, Alexander R. Steiner, Dan Groff, Juan Zhang, Alice Yam, Shengwen Liang, Trevor J. Hallam, Christopher D. Thanos, Joseph D. Kittle, Aaron K. Sato, Patrick Rivers, James Rozzelle, Junhao Yang, and Stephanie Armstrong
- Subjects
Protein Folding ,Saccharomyces cerevisiae Proteins ,Immunology ,Protein Disulfide-Isomerases ,Immunoglobulins ,Biology ,Antibodies, Monoclonal, Humanized ,Endoplasmic Reticulum ,Protein Engineering ,Immunoglobulin G ,Report ,Prolyl isomerase ,Humans ,Immunology and Allergy ,Protein disulfide-isomerase ,Bacteria ,Cell-Free System ,Escherichia coli Proteins ,Endoplasmic reticulum ,Protein engineering ,Periplasmic space ,Trastuzumab ,Recombinant Proteins ,Biochemistry ,Chaperone (protein) ,biology.protein ,Protein folding ,Biotechnology ,Molecular Chaperones - Abstract
Antibodies are well-established as therapeutics, and the preclinical and clinical pipeline of these important biologics is growing rapidly. Consequently, there is considerable interest in technologies to engineer and manufacture them. Mammalian cell culture is commonly used for production because eukaryotic expression systems have evolved complex and efficient chaperone systems for the folding of antibodies. However, given the ease and manipulability of bacteria, antibody discovery efforts often employ bacterial expression systems despite their limitations in generating high titers of functional antibody. Open-Cell Free Synthesis (OCFS) is a coupled transcription-translation system that has the advantages of prokaryotic systems while achieving high titers of antibody expression. Due to the open nature of OCFS, it is easily modified by chemical or protein additives to improve the folding of select proteins. As such, we undertook a protein additive screen to identify chaperone proteins that improve the folding and assembly of trastuzumab in OCFS. From the screen, we identified the disulfide isomerase DsbC and the prolyl isomerase FkpA as important positive effectors of IgG folding. These periplasmic chaperones function synergistically for the folding and assembly of IgG, and, when present in sufficient quantities, gram per liter IgG titers can be produced. This technological advancement allows the rapid development and manufacturing of immunoglobulin proteins and pushes OCFS to the forefront of production technologies for biologics.
- Published
- 2014
- Full Text
- View/download PDF
28. Production of Site-Specific Antibody–Drug Conjugates Using Optimized Non-Natural Amino Acids in a Cell-Free Expression System
- Author
-
Alice Yam, Gang Yin, Patrick Rivers, Trevor J. Hallam, Tyler H. Heibeck, Nathan Uter, Mary Rose Madlansacay, Cuong Tran, Avinash Gill, Wenjin Yang, Sunil Bajad, Erik S. Zimmerman, Alexander R. Steiner, Xiaofan Li, Aaron K. Sato, Chris Murray, Kalyani Penta, Christopher D. Thanos, and Willie D. Wang
- Subjects
Drug ,Immunoconjugates ,Immunogen ,medicine.drug_class ,media_common.quotation_subject ,Biomedical Engineering ,Pharmaceutical Science ,Bioengineering ,Pharmacology ,Monoclonal antibody ,Cell Line ,Therapeutic index ,Pharmacokinetics ,Tandem Mass Spectrometry ,medicine ,Humans ,Amino Acids ,media_common ,Cell-Free System ,Chemistry ,Immunogenicity ,Organic Chemistry ,Small molecule ,High-Throughput Screening Assays ,body regions ,Biochemistry ,Chromatography, Liquid ,Biotechnology ,Conjugate - Abstract
Antibody-drug conjugates (ADCs) are a targeted chemotherapeutic currently at the cutting edge of oncology medicine. These hybrid molecules consist of a tumor antigen-specific antibody coupled to a chemotherapeutic small molecule. Through targeted delivery of potent cytotoxins, ADCs exhibit improved therapeutic index and enhanced efficacy relative to traditional chemotherapies and monoclonal antibody therapies. The currently FDA-approved ADCs, Kadcyla (Immunogen/Roche) and Adcetris (Seattle Genetics), are produced by conjugation to surface-exposed lysines, or partial disulfide reduction and conjugation to free cysteines, respectively. These stochastic modes of conjugation lead to heterogeneous drug products with varied numbers of drugs conjugated across several possible sites. As a consequence, the field has limited understanding of the relationships between the site and extent of drug loading and ADC attributes such as efficacy, safety, pharmacokinetics, and immunogenicity. A robust platform for rapid production of ADCs with defined and uniform sites of drug conjugation would enable such studies. We have established a cell-free protein expression system for production of antibody drug conjugates through site-specific incorporation of the optimized non-natural amino acid, para-azidomethyl-l-phenylalanine (pAMF). By using our cell-free protein synthesis platform to directly screen a library of aaRS variants, we have discovered a novel variant of the Methanococcus jannaschii tyrosyl tRNA synthetase (TyrRS), with a high activity and specificity toward pAMF. We demonstrate that site-specific incorporation of pAMF facilitates near complete conjugation of a DBCO-PEG-monomethyl auristatin (DBCO-PEG-MMAF) drug to the tumor-specific, Her2-binding IgG Trastuzumab using strain-promoted azide-alkyne cycloaddition (SPAAC) copper-free click chemistry. The resultant ADCs proved highly potent in in vitro cell cytotoxicity assays.
- Published
- 2014
- Full Text
- View/download PDF
29. Wnt pathway inhibition via the targeting of Frizzled receptors results in decreased growth and tumorigenicity of human tumors
- Author
-
Kellie Pickell, Andrew Lam, Lucas Donigan, Marcus Fischer, Satyajit Sujit Kumar Mitra, Fumiko Takada Axelrod, Hoang Tran, Aurélie Chaudhari, In-Kyung Park, Christopher J. Bond, Austin L. Gurney, Timothy Hoey, Sanjeev Satyal, Aaron K. Sato, John Lewicki, Jennifer Cain, Cecile Chartier, Michelle Stroud, Sasha Lazetic, Ann M. Kapoun, May Ji, Wan-Ching Yen, and Shirley Ma
- Subjects
Frizzled ,Blotting, Western ,Genetic Vectors ,Cell ,Antineoplastic Agents ,CHO Cells ,Biology ,Epitope ,Immunoglobulin Fab Fragments ,Cricetulus ,Peptide Library ,Cricetinae ,Neoplasms ,medicine ,Animals ,Humans ,Luciferases ,Receptor ,Wnt Signaling Pathway ,Multidisciplinary ,Lentivirus ,HEK 293 cells ,Wnt signaling pathway ,Antibodies, Monoclonal ,LRP6 ,Drug Synergism ,LRP5 ,Biological Sciences ,Immunohistochemistry ,Molecular biology ,Frizzled Receptors ,Cell biology ,HEK293 Cells ,medicine.anatomical_structure - Abstract
The Wnt/β-catenin pathway, which signals through the Frizzled (Fzd) receptor family and several coreceptors, has long been implicated in cancer. Here we demonstrate a therapeutic approach to targeting the Wnt pathway with a monoclonal antibody, OMP-18R5. This antibody, initially identified by binding to Frizzled 7, interacts with five Fzd receptors through a conserved epitope within the extracellular domain and blocks canonical Wnt signaling induced by multiple Wnt family members. In xenograft studies with minimally passaged human tumors, this antibody inhibits the growth of a range of tumor types, reduces tumor-initiating cell frequency, and exhibits synergistic activity with standard-of-care chemotherapeutic agents.
- Published
- 2012
- Full Text
- View/download PDF
30. Phage Libraries
- Author
-
Aaron K. Sato
- Subjects
Chemistry - Published
- 2012
- Full Text
- View/download PDF
31. Discovery of Fully Human Anti-MET Monoclonal Antibodies with Antitumor Activity against Colon Cancer Tumor Models In Vivo
- Author
-
Min Wang, Timothy Velilla, Timothy Hoey, Yarrow Madrona, Andrew Lam, Aaron K. Sato, May Ji, Edward Thein Htun van der Horst, Lawrence Chinn, and Hoang Tran
- Subjects
Cancer Research ,biology ,Cell growth ,medicine.drug_class ,Monoclonal antibody ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Molecular biology ,lcsh:RC254-282 ,Receptor tyrosine kinase ,KLF6 ,In vivo ,Cancer research ,medicine ,biology.protein ,Phosphorylation ,Hepatocyte growth factor ,Signal transduction ,medicine.drug - Abstract
The receptor tyrosine kinase MET is a major component controlling the invasive growth program in embryonic development and in invasive malignancies. The discovery of therapeutic antibodies against MET has been difficult, and antibodies that compete with hepatocyte growth factor (HGF) act as agonists. By applying phage technology and cell-based panning strategies, we discovered two fully human antibodies against MET (R13 and R28), which synergistically inhibit HGF binding to MET and elicit antibody-dependent cellular cytotoxicity. Cell-based phosphorylation assays demonstrate that R13 and R28 abrogate HGF-induced activation of MET, AKT1, ERK1/2, and HGF-induced migration and proliferation. FACS experiments suggest that the inhibitory effect is mediated by “locking” MET receptor in a state with R13, which then increases avidity of R28 for the extracellular domain of MET, thus blocking HGF binding without activating the receptor. In vivo studies demonstrate that the combination of R13/28 significantly inhibited tumor growth in various colon tumor xenograft models. Inhibition of tumor growth was associated with induction of hypoxia. Global gene expression analysis shows that inhibition of HGF/MET pathway significantly upregulated the tumor suppressors KLF6, CEACAM1, and BMP2, the negative regulator of phosphatidylinositol-3-OH-kinase PIK3IP1, and significantly suppressed SCF and SERPINE2, both enhancers of proliferation and invasiveness. Moreover, in an experimental metastasis model, R13/28 increased survival by preventing the recurrence of otherwise lethal lung metastases. Taken together, these results underscore the utility of a dual-antibody approach for targeting MET and possibly other receptor tyrosine kinases. Our approach could be expanded to drug discovery efforts against other cell surface proteins.
- Published
- 2009
32. Identification of an ADAMTS-4 Cleavage Motif Using Phage Display Leads to the Development of Fluorogenic Peptide Substrates and Reveals Matrilin-3 as a Novel Substrate
- Author
-
Olga V. Nemirovskiy, Marc D. Zack, Aida Abujoub, Malini Viswanathan, Anne-Marie Malfait, Joseph W. Leone, Min Liu, Richard Mazzarella, Robert L. Hills, Micky D. Tortorella, Gary J. Bassill, Arumugam Muruganandam, Elizabeth C. Arner, Kam Fok, Aaron K. Sato, and Daniel J. Sexton
- Subjects
Phage display ,medicine.medical_treatment ,Amino Acid Motifs ,Molecular Sequence Data ,Protein Array Analysis ,Glutamic Acid ,Peptide ,Cleavage (embryo) ,Biochemistry ,Substrate Specificity ,Inhibitory Concentration 50 ,Peptide Library ,medicine ,Matrilin Proteins ,Protein Isoforms ,Amino Acid Sequence ,Peptide library ,Molecular Biology ,Aggrecan ,chemistry.chemical_classification ,Extracellular Matrix Proteins ,Protease ,Sequence Homology, Amino Acid ,Chemistry ,ADAMTS ,Cell Biology ,Recombinant Proteins ,Amino acid ,ADAM Proteins ,Kinetics ,Spectrometry, Fluorescence ,ADAMTS4 Protein ,Peptides ,Procollagen N-Endopeptidase ,Sequence Alignment - Abstract
ADAMTS-4 and ADAMTS-5 are aggrecanases responsible for the breakdown of cartilage aggrecan in osteoarthritis. Multiple ADAMTS-4 cleavage sites have been described in several matrix proteins including aggrecan, versican, and brevican, but no concise predictive cleavage motif has been identified for this protease. By screening a 13-mer peptide library with a diversity of 10(8), we have identified the ADAMTS-4 cleavage motif E-(AFVLMY)-X(0,1)-(RK)-X(2,3)-(ST)-(VYIFWMLA), with Glu representing P1. Several 13-mer peptides containing this motif, including DVQEFRGVTAVIR and HNEFRQRETYMVF, were shown to be substrates for ADAMTS-4. These peptides were found to be specific substrates for ADAMTS-4 as they were not cleaved by ADAMTS-5. Modification of these peptides with donor (6-FAM) and acceptor (QSY-9) molecules resulted in the development of fluorescence-based substrates with a Km of approximately 35 microM. Furthermore, the role of Glu at P1 and Phe at P1' in binding and catalysis was studied by exploring substitution of these amino acids with the D-isomeric forms. Substitution of P1 with dGlu was tolerable for binding, but not catalysis, whereas substitution of P1' with dPhe precluded both binding and catalysis. Similarly, replacement of Glu with Asp at P1 abolished recognition and cleavage of the peptide. Finally, BLAST results of the ADAMTS-4 cleavage motif identified matrilin-3 as a new substrate for ADAMTS-4. When tested, recombinant ADAMTS-4 effectively cleaved intact matrilin-3 at the predicted motif at Glu435/Ala436 generating two species of 45 and 5 kDa.
- Published
- 2007
- Full Text
- View/download PDF
33. Therapeutic peptides: technological advances driving peptides into development
- Author
-
Malini Viswanathan, Rachel Baribault Kent, Aaron K. Sato, and Clive R. Wood
- Subjects
Drug ,medicine.drug_class ,media_common.quotation_subject ,Biomedical Engineering ,Bioengineering ,Peptide ,Computational biology ,Pharmacology ,Biology ,Protein Engineering ,Key issues ,Monoclonal antibody ,Drug Delivery Systems ,Drug Stability ,medicine ,Technology, Pharmaceutical ,media_common ,chemistry.chemical_classification ,Protein therapeutics ,Small molecule ,Amino acid ,chemistry ,Drug Design ,Peptides ,Peptide drug ,Biotechnology - Abstract
As potential therapeutics, peptides offer several advantages over small molecules (increased specificity) and antibodies (small size). Nevertheless, a number of key issues have hampered their use as drug candidates. A series of new technologies have recently been developed that allow peptides to be viable drug candidates in areas usually restricted to protein therapeutics, such as monoclonal antibodies. These include the development of various types of peptide-conjugates that have lower rates of clearance and hence the potential to increase the exposure of peptide drug candidates in chronic diseases. Structural additions have also been made to peptides, including the use of unnatural amino acids, mainchain modifications and other novel substitutions, which have helped to improve peptide stability and further their therapeutic potential.
- Published
- 2006
- Full Text
- View/download PDF
34. Discovery of human antibodies against the C5aR target using phage display technology
- Author
-
Lili Huang, Aaron K. Sato, Daniel T. Dransfield, Susan Townsend, Tony J. Fleming, and Meena Sachdeva
- Subjects
Phage display ,medicine.drug_class ,Molecular Sequence Data ,Monoclonal antibody ,Immunoglobulin G ,Immunoglobulin Fab Fragments ,Peptide Library ,Structural Biology ,medicine ,Humans ,Amino Acid Sequence ,Peptide library ,Receptor, Anaphylatoxin C5a ,Molecular Biology ,Peptide sequence ,biology ,Membrane Proteins ,Cell Differentiation ,U937 Cells ,Molecular biology ,Receptors, Complement ,Biochemistry ,Biotinylation ,biology.protein ,Antibody - Abstract
Phage display technologies have been increasingly utilized for the generation of therapeutic, imaging and purification reagents for a number of biological targets. Using a variety of different approaches, we have developed antibodies with high specificity and affinity for various targets ranging from small peptides to large proteins, soluble or membrane-associated as well as to activated forms of enzymes. We have applied this approach to G-protein coupled receptors (GPCRs), often considered difficult targets for antibody therapeutics and targeting. Here we demonstrate the use of this technology for the identification of human antibodies targeting C5aR, the chemoattractant GPCR receptor for anaphylatoxin C5a. The N-terminal region (residues 1-31) of C5aR, one of the ligand binding sites, was synthesized, biotinylated and used as the target for selection. Three rounds of selection with our proprietary human Fab phage display library were performed. Screening of 768 isolates by phage ELISA identified 374 positive clones. Based on sequence alignment analysis, the positive clones were divided into 22 groups. Representative Fab clones from each group were reformatted into IgGs and tested for binding to C5aR-expressing cells, the differentiated U-937 cells. Flow cytometric analysis demonstrated that nine out of 16 reformatted IgGs bound to cells. Competition with a C5aR monoclonal antibody S5/1 which recognizes the same N-terminal region showed that S5/1 blocked the binding of positive cell binders to the peptide used for selections, indicating that the identified cell binding IgGs were specific to C5aR. These antibody binders represent viable candidates as therapeutic or imaging agents, illustrating that phage display technology provides a rapid means for developing antibodies to a difficult class of targets such as GPCRs.
- Published
- 2005
- Full Text
- View/download PDF
35. Production of bispecific antibodies in 'knobs-into-holes' using a cell-free expression system
- Author
-
Junhao Yang, Cuong Tran, Willie D. Wang, Trevor J. Hallam, Gang Yin, Aaron K. Sato, Tyler H. Heibeck, Ryan Stafford, Alexander R. Steiner, John Lee, and Yiren Xu
- Subjects
Dual target ,Bispecific antibody ,Immunology ,Gene Expression ,Cell free ,Computational biology ,CHO Cells ,cell-free protein expression ,scFv-KIH ,Cricetulus ,Cricetinae ,Antibodies, Bispecific ,prefabrication ,Immunology and Allergy ,Animals ,Humans ,BiTE-KIH ,Heavy chain ,Cell-Free System ,Chemistry ,Target engagement ,Molecular biology ,Recombinant Proteins ,bispecific antibody ,ELISA - Enzyme-linked immunosorbent assay ,knob-into-hole ,Single-Chain Antibodies ,Reports - Abstract
Bispecific antibodies have emerged in recent years as a promising field of research for therapies in oncology, inflammable diseases, and infectious diseases. Their capability of dual target recognition allows for novel therapeutic hypothesis to be tested, where traditional mono-specific antibodies would lack the needed mode of target engagement. Among extremely diverse architectures of bispecific antibodies, knobs-into-holes (KIHs) technology, which involves engineering CH3 domains to create either a "knob" or a "hole" in each heavy chain to promote heterodimerization, has been widely applied. Here, we describe the use of a cell-free expression system (Xpress CF) to produce KIH bispecific antibodies in multiple scaffolds, including 2-armed heterodimeric scFv-KIH and one-armed asymmetric BiTE-KIH with tandem scFv. Efficient KIH production can be achieved by manipulating the plasmid ratio between knob and hole, and further improved by addition of prefabricated knob or hole. These studies demonstrate the versatility of Xpress CF in KIH production and provide valuable insights into KIH construct design for better assembly and expression titer.
- Published
- 2014
36. Determinants of the Peptide-induced Conformational Change in the Human Class II Major Histocompatibility Complex Protein HLA-DR1
- Author
-
George B. Benedek, Scheherazade Sadegh-Nasseri, Sateesh Kumar Natarajan, Mia M. Rushe, Aleksey Lomakin, Jennifer A. Zarutskie, Lawrence J. Stern, and Aaron K. Sato
- Subjects
Models, Molecular ,Protein Folding ,Conformational change ,Protein Conformation ,Stereochemistry ,HLA-DR1 ,Molecular Sequence Data ,Mutant ,Enzyme-Linked Immunosorbent Assay ,Peptide ,Biology ,Biochemistry ,Antigen ,Side chain ,Humans ,Point Mutation ,Scattering, Radiation ,Amino Acid Sequence ,skin and connective tissue diseases ,Molecular Biology ,chemistry.chemical_classification ,Dose-Response Relationship, Drug ,Circular Dichroism ,HLA-DR1 Antigen ,Histocompatibility Antigens Class II ,Temperature ,Class II major histocompatibility complex protein ,Cell Biology ,Kinetics ,chemistry ,Mutagenesis ,Chromatography, Gel ,Thermodynamics ,sense organs ,Peptides ,Intracellular ,Protein Binding - Abstract
The human class II major histocompatibility complex protein HLA-DR1 has been shown previously to undergo a distinct conformational change from an open to a compact form upon binding peptide. To investigate the role of peptide in triggering the conformational change, the minimal requirements for inducing the compact conformation were determined. Peptides as short as two and four residues, which occupy only a small fraction of the peptide-binding cleft, were able to induce the conformational change. A mutant HLA-DR1 protein with a substitution in the beta subunit designed to fill the P1 pocket from within the protein (Gly(86) to Tyr) adopted to a large extent the compact, peptide-bound conformation. Interactions important in stabilizing the compact conformation are shown to be distinct from those responsible for high affinity binding or for stabilization of the complex against thermal denaturation. The results suggest that occupancy of the P1 pocket is responsible for partial conversion to the compact form but that both side chain and main chain interactions contribute to the full conformational change. The implications of the conformational change to intracellular antigen loading and presentation are discussed.
- Published
- 2000
- Full Text
- View/download PDF
37. Extracellular antigen processing and presentation by immature dendritic cells
- Author
-
Gregory J. Carven, Laura Santambrogio, Jack L. Strominger, Svetlana L. Belyanskaya, Aaron K. Sato, and Lawrence J. Stern
- Subjects
Antigen Presentation ,HLA-D Antigens ,Multidisciplinary ,biology ,Immunodominant Epitopes ,Antigen processing ,Antigen presentation ,Histocompatibility Antigens Class II ,Dendritic Cells ,HLA-DM ,Biological Sciences ,MHC restriction ,Major histocompatibility complex ,Peptide Fragments ,Cell biology ,Antigens, Differentiation, B-Lymphocyte ,Mice ,Antigen ,MHC class I ,biology.protein ,Animals ,Humans ,Cytotoxic T cell ,Protein Binding - Abstract
In antigen presentation to CD4+T cells, proteins are degraded to peptide fragments and loaded onto class II MHC molecules in a process involving the peptide exchange factors H-2M (murine) or HLA-DM (human). In many antigen-presenting cells these processes occur in intracellular endosomal compartments, where peptides are generated and loaded onto class II MHC proteins for subsequent transport to the surface and presentation to T cells. Here, we provide evidence for an additional antigen-processing pathway in immature dendritic cells (DC). Immature DC express at the cell surface empty or peptide-receptive class II MHC molecules, as well as H-2M or HLA-DM. Secreted DC proteases act extracellularly to process intact proteins into antigenic peptides. Peptides produced by such activity are efficiently loaded onto cell surface class II MHC molecules. Together these elements comprise an unusual extracellular presentation pathway in which antigen processing and peptide loading can occur entirely outside of the cell.
- Published
- 1999
- Full Text
- View/download PDF
38. Abundant empty class II MHC molecules on the surface of immature dendritic cells
- Author
-
Laura Santambrogio, Aaron K. Sato, Lawrence J. Stern, Martin E. Dorf, and Falko R. Fischer
- Subjects
T-Lymphocytes ,Antigen presentation ,CD1 ,Bone Marrow Cells ,Lymphocyte Activation ,Major histocompatibility complex ,Mice ,Antigen ,Bone Marrow ,MHC class I ,Animals ,Humans ,Antigens ,HLA-D Antigens ,Antigen Presentation ,Multidisciplinary ,biology ,Antigen processing ,HLA-DR1 Antigen ,Histocompatibility Antigens Class II ,Dendritic Cells ,Biological Sciences ,MHC restriction ,Molecular biology ,Recombinant Proteins ,biology.protein ,Spleen ,Protein Binding - Abstract
A monoclonal antibody specific for the empty conformation of class II MHC molecules revealed the presence of abundant empty molecules on the surface of spleen- and bone marrow-derived dendritic cells (DC) among various types of antigen-presenting cells. The empty class II MHC molecules are developmentally regulated and expressed predominantly on immature DC. They can capture peptide antigens directly from the extracellular medium and present bound peptides to antigen-specific T lymphocytes. The ability of the empty cell-surface class II MHC proteins to bind peptides and present them to T cells without intracellular processing can serve to extend the spectrum of antigens able to be presented by DC, consistent with their role as sentinels in the immune system.
- Published
- 1999
- Full Text
- View/download PDF
39. Substitution of aspartic acid at β57 with alanine alters MHC class II peptide binding activity but not protein stability: HLA-DQ (α1∗0201, β1∗0302) and (α1∗0201, β1∗0303)
- Author
-
Tiziana Sturniolo, Aaron K. Sato, Francesco Sinigaglia, and Lawrence J. Stern
- Subjects
Alanine ,Aspartic Acid ,MHC class II ,biology ,Chemistry ,Immunology ,Peptide binding ,General Medicine ,Major histocompatibility complex ,Protein Structure, Secondary ,Protein Structure, Tertiary ,Serine ,Amino Acid Substitution ,Solubility ,Biochemistry ,Valine ,HLA-DQ Antigens ,HLA-DQ ,Aspartic acid ,biology.protein ,Humans ,Immunology and Allergy ,Peptides - Abstract
In class II major histocompatibility complex (MHC) proteins, residue beta57 is usually aspartic acid. Alleles carrying serine, valine, or alanine at this position are strongly correlated with the development of insulin-dependent diabetes mellitus (IDDM). Asp(beta)57 participates in a conserved salt bridge that bridges the alpha and beta subunits in the peptide-binding site. It has been proposed that the correlation between IDDM and MHC alleles lacking Asp(beta)57 may be due to an instability of the protein caused by loss of this salt bridge. Using a pair of HLA-DQ proteins (alpha1*0201, beta1*0302) and (alpha1*0201, beta1*0303) differing only in having aspartic acid or alanine at position beta57, we show that the polymorphism does not have a significant effect on protein stability for either the empty or peptide-loaded forms. However, the circular dichroism spectra indicate that empty and peptide-loaded Alabeta57 proteins display slightly different secondary structures relative to their Aspbeta57 counterparts. A set of three peptides shows different binding affinities for DQ(alpha1*0201, beta1*0302) relative to DQ(alpha1*0201, beta1*0303). We propose that substitution of Asp(beta)57 residue causes a local rearrangement within the DQ peptide-binding site that alters the peptide-binding specificity. This rearrangement may help to explain the previously observed differences in SDS stability between Asp and non-Asp(beta)57 DQ proteins.
- Published
- 1999
- Full Text
- View/download PDF
40. A Conformational Change in the Human Major Histocompatibility Complex Protein HLA-DR1 Induced by Peptide Binding
- Author
-
Aleksey Lomakin, George B. Benedek, Lawrence J. Stern, Iat C. Chan, Jennifer A. Zarutskie, Mia M. Rushe, and Aaron K. Sato
- Subjects
Models, Molecular ,Protein Folding ,Circular dichroism ,Conformational change ,Protein Conformation ,Molecular Sequence Data ,Antigen presentation ,Peptide binding ,Cooperativity ,Peptide ,Major histocompatibility complex ,Biochemistry ,Protein Structure, Secondary ,Humans ,Amino Acid Sequence ,skin and connective tissue diseases ,chemistry.chemical_classification ,biology ,Chemistry ,Circular Dichroism ,HLA-DR1 Antigen ,Antibodies, Monoclonal ,Hydrogen-Ion Concentration ,Recombinant Proteins ,Kinetics ,biology.protein ,Thermodynamics ,sense organs ,Peptides ,Intracellular ,Protein Binding - Abstract
To investigate a conformational change accompanying peptide binding to class II MHC proteins, we probed the structure of a soluble version of the human class II MHC protein HLA-DR1 in empty and peptide-loaded forms. Peptide binding induced a large decrease in the effective radius of the protein as determined by gel filtration, dynamic light scattering, and analytical ultracentrifugation. It caused a substantial increase in the cooperativity of thermal denaturation and induced alterations in MHC polypeptide backbone structure as determined by circular dichroism. These changes suggest a condensation of the protein around the bound peptide. An antibody specific for beta58-69 preferentially bound the empty protein, indicating that the peptide-induced conformational change involves the beta-subunit helical region. The conformational change may have important implications for the mechanisms of intracellular antigen presentation pathways.
- Published
- 1999
- Full Text
- View/download PDF
41. Empty and Peptide-Loaded Class II Major Histocompatibility Complex Proteins Produced by Expression inEscherichia coliand Foldingin Vitro
- Author
-
Lawrence J. Stern, Mia Frayser, Aaron K. Sato, and Lihui Xu
- Subjects
Protein Denaturation ,Protein Folding ,Hot Temperature ,Macromolecular Substances ,Protein Conformation ,Proteolysis ,Molecular Sequence Data ,Peptide ,medicine.disease_cause ,Antibodies ,law.invention ,Protein structure ,law ,Escherichia coli ,medicine ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Binding site ,Peptide sequence ,chemistry.chemical_classification ,Binding Sites ,medicine.diagnostic_test ,Chemistry ,HLA-DR1 Antigen ,Peptide Fragments ,Recombinant Proteins ,Kinetics ,Biochemistry ,Recombinant DNA ,Thermodynamics ,Protein folding ,Biotechnology - Abstract
The human class II major histocompatibility complex protein HLA-DR1 has been expressed in Escherichia coli as denatured alpha and beta subunits and folded in vitro to form the native structure. DR1 folding yields are 30-50% in the presence or absence of tight-binding antigenic peptides. The protein produced in this manner is soluble and monomeric with the expected apparent molecular weight. It reacts with conformation-sensitive anti-DR antibodies and exhibits peptide-dependent resistance to SDS-induced chain dissociation and to proteolysis as does the native protein. The observed peptide specificity and dissociation kinetics are similar to those of native DR produced in B-cells and finally the protein exhibits circular dichroism spectra and cooperative thermal denaturation as expected for a folded protein. We conclude that the recombinant DR1 has adopted the native fold. We have folded DR1 in the absence of peptide and isolated a soluble, peptide-free alphabeta-heterodimer. The empty DR1 can bind antigenic peptide but exhibits altered far UV-circular dichroism and thermal denaturation relative to the peptide-bound form.
- Published
- 1999
- Full Text
- View/download PDF
42. Discovery and Preclinical Development of Novel CD74-Targeting Antibody-Drug Conjugates (ADCs) with Significant Activity in Multiple Myeloma (MM) Cell Lines and Xenograft Models
- Author
-
Alexander Steiner, Shannon Matheny, John F. Boylan, Ryan Stafford, Heather Stephenson, Cristina Abrahams, Alice Yam, Arturo Molina, Stuart Bussell, Henry Heinsohn, Aaron K. Sato, Maureen Fitch Bruhns, Abigail Yu, Heidi Hoffmann, Adam Galan, Nicki Vasquez, Eric Schwartz, Stellanie Krimm, Venita DeAlmeida, James Zawada, Jason Kahana, Trevor J. Hallam, Mark Lupher, Toni Kline, Rama K. Narla, and Xiaofan Li
- Subjects
0301 basic medicine ,Drug ,CD74 ,biology ,business.industry ,media_common.quotation_subject ,Immunology ,Cell Biology ,Hematology ,Pharmacology ,medicine.disease ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,Cell killing ,Cell culture ,biology.protein ,Potency ,Medicine ,Antibody ,business ,Multiple myeloma ,Conjugate ,media_common - Abstract
CD74, also known as HLA-DR-associated invariant chain, is a type II transmembrane glycoprotein highly expressed in many B-cell malignancies. The limited expression of CD74 in normal tissues suggests it may be a suitable ADC target for these tumor types. Accordingly, we engineered an anti-CD74 human IgG1 antibody (SP7219) using novel Fab-based ribosome display methods. The selected Fabs were readily reformatted and directly screened as IgGs using Sutro's unique high-throughput, cell-free protein synthesis platform, Xpress CFTM. We then developed novel, potent ADCs, SP7676 and SP7675 (STRO-001), comprised of our lead antibody (SP7219) conjugated to non-cleavable DBCO-maytansinoid linker-warheads with an average drug-antibody ratios (DAR) of 2. We used site-specific conjugation technology which results in a high degree of homogeneity characterized by the drug linker covalently binding to a single defined site. The sites for conjugation were selected based on highest cell killing activity and stablity in vitro and in vivo. Both ADCs demonstrate potent cell killing activity across multiple B-cell tumor lines in vitro, and anti-tumor activity in preclinical multiple myeloma xenograft models. In vitro cytotoxicity assays show nanomolar potency of STRO-001 in four MM cell lines: Mc/CAR (IC50 0.8 nM), MM.1S (IC50 10-11 nM), U266B1 (IC50 8.5 -9.3 nM), and ARP-1 (IC50 4.3-22 nM). CD74 cell surface expression is required for ADC anti-proliferative effect but the expression level does not seem to correlate with in vitro potency. SP7676 elicited a robust anti-tumor response in the ANBL-6 multiple myeloma xenograft model. Durable regressions were observed in all mice at ≥ 3 mg/kg, with equivalent efficacy (regression) at 3 mg/kg (every 3 days x5) and 10 mg/kg (every 3 days x5 or weekly x3). SP7676 also elicited a clear survival benefit in a disseminated multiple myeloma CAG xenograft model starting at 1mg/kg every 3 days x 5 doses. Similarly, both SP7676 and STRO-001 inhibited the formation of internal visceral tumors in the ARP-1 xenograft model after 3 weekly doses of 3 mg/kg. Evaluation of our lead candidate, STRO-001 in additional MM cell lines and primary patient samples is planned. The tolerability of STRO-001 in non-human primates is under evaluation. STRO-001 was administered to cynomolgous monkeys in an exploratory dose-escalating study up to 30 mg/kg x 2 doses on Day 1 and 15. STRO-001 reduces normal B-cell populations at ≥1 mg/kg after a single dose, providing pharmacodynamic evidence of B-cell targeting while other hematopoietic lineages are mostly affected only at the highest dose studied. Anticipated hematologic toxicities were readily reversible at 1, 3 and 10 mg/kg and target organs of interest were identified. Based on these encouraging data, STRO-001 is advancing to IND-enabling studies for the treatment of CD74 expressing multiple myeloma and other B-cell malignancies. Disclosures Abrahams: Sutro Biopharma: Employment. Li:Sutro Biopharma: Employment. Yu:Sutro Biopharma: Employment. Krimm:Sutro Biopharma: Employment. Kahana:Celgene: Employment. Narla:Celgene: Employment. Schwartz:Celgene: Employment. Boylan:Celgene: Employment. Hoffmann:Sutro Biopharma: Employment. Steiner:Sutro Biopharma: Employment. Zawada:Sutro Biopharma: Employment. Stephenson:Sutro Biopharma: Employment. Bruhns:Sutro Biopharma: Employment. DeAlmeida:Sutro Biopharma: Employment. Matheny:Sutro Biopharma: Employment. Bussell:Sutro Biopharma: Employment. Galan:Sutro Biopharma: Employment. Kline:Sutro Biopharma: Employment. Vasquez:Sutro Biopharma: Employment. Yam:Sutro Biopharma: Employment. Stafford:Sutro Biopharma: Employment. Heinsohn:Sutro Biopharma: Employment. Sato:Sutro Biopharma: Employment. Molina:Sutro Biopharma: Employment. Hallam:Sutro Biopharma: Employment. Lupher:Sutro Biopharma: Employment.
- Published
- 2016
- Full Text
- View/download PDF
43. Targeting CD74 with Novel Antibody Drug Conjugates (ADCs) for the Treatment of B-Cell Non-Hodgkin's Lymphoma (NHL)
- Author
-
Alice Yam, Ryan Stafford, Maureen Fitch Bruhns, Millicent Embry, Stuart Bussell, Arturo Molina, Trevor J. Hallam, Mark Lupher, Toni Kline, Adam Galan, Shannon Matheny, Michael Brown, Venita DeAlmeida, John F. Boylan, Eric Schwartz, Cristina Abrahams, Jason Kahana, Alexander Steiner, Abigail Yu, Aaron K. Sato, James Zawada, Heather Stephenson, Leo Barnes, Henry Heinsohn, Nicki Vasquez, Rama K. Narla, Heidi Hoffmann, and Xiaofan Li
- Subjects
0301 basic medicine ,business.industry ,Chronic lymphocytic leukemia ,Immunology ,Follicular lymphoma ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Non-Hodgkin's lymphoma ,Lymphoma ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Cell killing ,medicine ,Cancer research ,Mantle cell lymphoma ,business ,Diffuse large B-cell lymphoma ,B cell - Abstract
CD74 is a type II transmembrane glycoprotein involved in the formation and transport of MHC class II protein. High expression of CD74 has been confirmed in follicular lymphoma, diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL) and other types of NHL with immunohistochemistry (IHC) using the LL1 antibody (Stein et al. Clin Cancer Res 2007). We employed site-specific conjugation technology to generate novel CD74-targeting ADCs, SP7676 and SP7675 (STRO-001) that exhibit a high degree of homogeneity characterized by the drug linker covalently binding to a single defined site. The human anti-CD74 IgG1 antibody (SP7219) used for ADCs SP7676 and STRO-001 was engineered using novel Fab-based ribosome display methods enabling selection from ~1012 different antibody variants. Hundreds of unique Fabs from this selection were converted to IgGs and expressed directly in Sutro's proprietary cell-free protein synthesis platform, Xpress CFTM, for extensive screening. The top antibody lead derived from this screen is further tested to identify the best sites for conjugation of linker-warheads. Sutro's SP7219 emerged as the top performing antibody and was conjugated to noncleavable DBCO-maytansinoid linker-warheads to form the ADCs SP7676 and STRO-001. Since conjugation sites were selected based on highest stability both in vitro and in vivo, these novel ADCs lose little drug moiety in circulation and have potential for improved PK, safety and activity profiles. In vitro cell proliferation/cytotoxicity assays show potent activity in 1) DLBCL (germinal center B-cell-like [GCB] and "double-hit") lines: SU-DHL-4, IC50 - 1nM; SU-DHL-6, IC50 - 0.4 nM; WSU-NHL, IC50 - 1.6 nM; Pfeiffer, IC50 - .09 nM; NUDUL-1, IC50 - 0.4 nM; HT, IC50 - 0.7 nM; OCI-LY-19, IC50 - 0.7 nM; WSU-DLBCL2, IC50 - 0.3 nM; 2) mantle cell lymphoma (MCL) cell lines: Mino, IC50 - 0.4-0.7 nM; JVM-2, IC50 - 1.7-2.9 nM; Jeko-1, IC50 - 0.4 - 0.6 nM; 3) Ph+ acute lymphoblastic leukemia (ALL): SUP-B15, IC50 - 3.9-4.6 nM; and 4) CLL (EBV-transformed): JVM-13, IC50 - 3.0-3.4 nM. SP7676 elicited strong anti-tumor response in the OCI-LY-10 lymphoma xenograft model with 100% of animals achieving complete regression of tumors at 3mg/kg every 3 days x 5 doses and 10 mg/kg weekly x 3 doses. In the WSU-DLCL2 "double-hit" lymphoma xenograft model, administration of SP7676 (with re-dosing at time of re-growth) produced tumor regressions at 10 mg/kg every 3 days x 5 (6/8 mice tumor free, remaining 2 with small tumors) and 10 mg/kg weekly x 3 (tumor regression in most animals, 4/8 tumor free). Additionally, STRO-001 exhibits dose-dependent tumor growth inhibition in SUDHL-6 xenografts starting at 2.5 mg/kg weekly x 3 doses. Exploratory testing of our lead candidate, STRO-001 in cynomologous monkeys showed dose-dependent B-cell depletion at 1 - 30 mg/kg doses on Day 1 and 15, confirming the intended pharmacodynamic effect. Our preliminary data demonstrate that SP7676 and STRO-001 generate potent cell killing activity across multiple B-cell lymphoma/leukemia cell lines in vitro, and anti-tumor activity in preclinical B-cell NHL xenografts. Evaluation of STRO-001 in other cell lines and xenograft models and in combination studies is ongoing. GLP toxicology and other IND-enabling studies are planned. Disclosures Li: Sutro Biopharma: Employment. Abrahams:Sutro Biopharma: Employment. Embry:Sutro Biopharma: Employment. Yu:Sutro Biopharma: Employment. Kahana:Celgene: Employment. Brown:Celgene: Employment. Narla:Celgene: Employment. Barnes:Celgene: Employment. Schwartz:Celgene: Employment. Boylan:Celgene: Employment. Zawada:Sutro Biopharma: Employment. Stephenson:Sutro Biopharma: Employment. Bruhns:Sutro Biopharma: Employment. Bussell:Sutro Biopharma: Employment. Steiner:Sutro Biopharma: Employment. Galan:Sutro Biopharma: Employment. Kline:Sutro Biopharma: Employment. Yam:Sutro Biopharma: Employment. Stafford:Sutro Biopharma: Employment. Hoffmann:Sutro Biopharma: Employment. Matheny:Sutro Biopharma: Employment. DeAlmeida:Sutro Biopharma: Employment. Vasquez:Sutro Biopharma: Employment. Heinsohn:Sutro Biopharma: Employment. Sato:Sutro Biopharma: Employment. Molina:Sutro Biopharma: Employment. Hallam:Sutro Biopharma: Employment. Lupher:Sutro Biopharma: Employment.
- Published
- 2016
- Full Text
- View/download PDF
44. Abstract A41: Use of patient-derived tumor xenografts (PDX) for discovery and development of an anti-Notch2/3 monoclonal antibody targeting cancer stem cells
- Author
-
John Lewicki, Jalpa Shaw, Tim Hoey, Marcus Fischer, Fumiko Takada Axelrod, Wan-Ching Yen, Christopher J. Bond, Austin L. Gurney, Jakob Dupont, Belinda Cancilla, Min Wang, Aaron K. Sato, Chun Zhang, Breanna Wallace, Lei Zhou, Jennifer Cain, Rene Meisner, Ann M. Kapoun, Randall Henner, and Tracy Tang
- Subjects
Cancer Research ,education.field_of_study ,biology ,business.industry ,CD44 ,Population ,Notch signaling pathway ,Cancer ,medicine.disease ,Gemcitabine ,Oncology ,Cancer stem cell ,Immunology ,Cancer research ,medicine ,biology.protein ,Tarextumab ,Stem cell ,business ,education ,medicine.drug - Abstract
Accumulating evidence has suggested that tumors may arise and grow as a result of the formation of a subset cell population termed cancer stem cells (CSC) or tumor initiating cells. Several research reports have indicated that CSCs are relatively resistant to conventional therapies. Thus, therapeutic strategies that specifically target cancer stem cells could have a major impact on cancer patient survival. Patient-derived tumor xenografts (PDX) have played a major role in the development of new cancer therapies. The advantage of PDX over standard cell-line xenograft models is that PDX retain much of the molecular, genetic, and histological heterogeneity of the original tumor and are minimally passed. In addition, self-renewal and lineage differentiation, the hallmarks of cancer stem cells, can be demonstrated through serial transplantation in immunodeficient mice. Our therapeutic approach in cancer stem cell drug discovery has been to target key developmental pathways that have been strongly implicated in cancer including the Notch pathway. We carried out tumorigenicity studies in three SCLC PDX models utilizing naïve cells positively enriched for either Notch2 or Notch3 expression. In two of the three models, Notch2+ and/or Notch3+ cell populations resulted in enhanced tumor formation. In a SCLC tumor that expressed low levels of Notch3, the Notch2+ population resulted in 70% tumor formation compared to CD44+ (10%) or CD133+ (45%) enriched populations. We have developed a monoclonal antibody, tarextumab (OMP-59R5), which selectively inhibits the function of both Notch2 and Notch3. Our preclinical data in PDX models demonstrate that tarextumab was efficacious in inhibiting the growth of various indications with minimal intestinal toxicity. Notably, the sensitivity of tarextumab in combination with gemcitabine or gemcitabine plus nab-paclitaxel in pancreatic tumors was associated with higher levels of Notch3 gene expression. Interference with Notch2/3 signaling by tarextumab delays tumor recurrence, decreases cancer stem cell frequency (as determined by in vivo LDA studies) and modulates the function of tumor vasculature. Our ALPINE Phase 1b clinical trial indicates that tarextumab is generally well-tolerated and shows signs of anti-tumor efficacy and modulation of Notch pathway signaling in the clinic. Furthermore, we observe a higher response rate and longer survival in patients with Notch3 high tumors receiving gemcitabine/nab-paclitaxel/tarextumab combination therapy. Analysis of pre- and post-treatment tumor biopsies showed an inhibition of Notch pathway and CSC gene signatures. Ongoing Phase 2 clinical trials evaluate first-line treatment with tarextumab in metastatic pancreatic cancer (ALPINE) and small cell lung cancer (PINNACLE). Collectively, these results demonstrate the utility of PDX models for discovery and development of anti-cancer stem cell therapeutics, identifying pharmacodynamic endpoints of drug actions, identifying predictive biomarkers for patient stratification, and translating these preclinical findings into clinical trials. Citation Format: Marcus M. Fischer, Wan-Ching Yen, Fumiko Axelrod, Christopher Bond, Jennifer Cain, Belinda Cancilla, Randall Henner, Rene Meisner, Aaron Sato, Jalpa Shaw, Tracy Tang, Breanna Wallace, Min Wang, Chun Zhang, Ann Kapoun, Lei Zhou, Jakob Dupont, John Lewicki, Austin Gurney, Tim Hoey. Use of patient-derived tumor xenografts (PDX) for discovery and development of an anti-Notch2/3 monoclonal antibody targeting cancer stem cells. [abstract]. In: Proceedings of the AACR Special Conference: Patient-Derived Cancer Models: Present and Future Applications from Basic Science to the Clinic; Feb 11-14, 2016; New Orleans, LA. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(16_Suppl):Abstract nr A41.
- Published
- 2016
- Full Text
- View/download PDF
45. Abstract B44: Targeting Notch signaling with a dual Notch 2/3 antagonist, OMP-59R5, inhibits tumor growth and decreases cancer stem cell frequency in pancreatic cancer
- Author
-
Marcus Fischer, Ann M. Kapoun, Timothy Hoey, John Lewicki, Austin L. Gurney, Min Wang, Wan-Ching Yen, and Aaron K. Sato
- Subjects
business.industry ,Notch signaling pathway ,Cancer ,medicine.disease ,Gemcitabine ,Pancreatic tumor ,Cancer stem cell ,Pancreatic cancer ,Cancer cell ,medicine ,Cancer research ,business ,Notch 2 ,medicine.drug - Abstract
Clinical management of pancreatic cancer is challenging and in recent years has not been dramatically improved and this disease remains the fourth most common cause of cancer-related death. Dysregulated Notch2 and/or Notch3 activity has been associated with several human tumor types including pancreatic cancer. In this report, we describe the development of OMP-59R5, an antibody isolated originally by binding to Notch2 and subsequently found to also bind Notch3. OMP-59R5 potently blocks Notch2 and Notch3 signaling and inhibits the growth pancreatic cancer patient derived xenograft tumors, as a single agent and in combination with gemcitabine. The anti-tumor effects were associated with modulation of Notch target genes in tumor and stromal cells. Inclusion of OMP-59R5 delayed pancreatic tumor recurrence following termination of gemcitabine, correlated with an effect of anti-Notch2/3 in decreasing in cancer stem cell frequency and tumorigenicity. Gene set enrichment analysis showed that a subset of the stem cell gene sets and gene set related to epithelial-to-mesenchymal transistion, EMT, was up-regulated in tumors by gemcitabine alone, but down-regulated by the combination of gemcitabine plus OMP-59R5 treatment. Findings from the present study suggest that agents that reduce cancer cell frequency may improve cancer treatment by delaying or preventing tumor recurrence, and reducing the metastatic spread of the disease. In particular, blocking Notch-mediated proliferation of bulk tumor cells and cancer stem cells may provide an attractive strategy for novel therapy in pancreatic cancer treatment. Citation Format: Wan-Ching Yen, Marcus Fischer, Aaron Sato, Min Wang, Ann M. Kapoun, John Lewicki, Austin Gurney, Timothy Hoey. Targeting Notch signaling with a dual Notch 2/3 antagonist, OMP-59R5, inhibits tumor growth and decreases cancer stem cell frequency in pancreatic cancer. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Progress and Challenges; Jun 18-21, 2012; Lake Tahoe, NV. Philadelphia (PA): AACR; Cancer Res 2012;72(12 Suppl):Abstract nr B44.
- Published
- 2012
- Full Text
- View/download PDF
46. Steady-state and time-resolved phosphorescence of wild-type and modified bacteriophage λcI repressors
- Author
-
Eric R. Bitten, Aaron K. Sato, J. B. Alexander Ross, Kenneth W. Rousslang, and Donald F. Senear
- Subjects
Sociology and Political Science ,biology ,Chemistry ,Clinical Biochemistry ,Tryptophan ,Wild type ,Repressor ,Photochemistry ,biology.organism_classification ,Biochemistry ,Bacteriophage ,Absorbance ,Clinical Psychology ,Excited state ,Steady state (chemistry) ,Phosphorescence ,Law ,Spectroscopy ,Social Sciences (miscellaneous) - Abstract
We have measured the steady-state phosphorescence and decay times of wild-type λcI repressor and compared it with that of a modified λcI repressor in which > 95% of the tryptophans were replaced with 5-hydroxy-l-tryptophan (5-OHTrp). The wild-type and 5-OHTrp-λcI repressors are spectroscopically distinct such that we can selectively excite the 5-OHTrp-λcI even in the presence of a 15-fold molar excess ofN-acetyltryptophanamide (NATrpA). The phosphorescence band of wild-type λcI is red-shifted by 3 nm relative to NATrpA, characteristic of buried tryptophan. Similarly, the phosphorescence of 5-OHTrp-λcI repressor is red-shifted relative to the model, 5-OHTrp, showing that according to the phosphorescence, the modified repressor is structurally indistinguishable from the native repressor. While the phosphorescence decay of both NATrpA and 5-OHTrp are single exponentials, the decay of both wild-type and 5-OHTrp-λcI repressors is complex, requiring three decay components whose fractional contributions to the phosphorescence are the same for both repressors. Because the 5-OHTrp phosphorescence can be excited at wavelengths outside the absorbance range of tryptophan and DNA, a protein spectrally enhanced with this emitter will aid the investigations of protein-protein or protein-DNA interactions.
- Published
- 1994
- Full Text
- View/download PDF
47. Abstract A33: Combined therapy of a novel anti-NOTCH2/3 antibody with paclitaxel inhibits tumor growth in a patient-derived breast tumor xenograft
- Author
-
Christopher Murriel, Austin L. Gurney, Breanna Wallace, John Lewicki, Jalpa Shah, Jennifer Cain, Tim Hoey, Aaron K. Sato, Jie Wei, Ann M. Kapoun, Tracy Tang, Cancilla Belinda, and Min Wang
- Subjects
JAG2 ,Cancer Research ,JAG1 ,business.industry ,Notch signaling pathway ,Cancer ,Pharmacology ,medicine.disease ,Metastasis ,chemistry.chemical_compound ,Breast cancer ,Oncology ,Paclitaxel ,chemistry ,Cancer stem cell ,medicine ,Cancer research ,business - Abstract
Cancer stem cells (CSCs) have been identified as the small subpopulation of cells in many cancers and are thought to be responsible for cancer initiation, progression, metastasis, recurrence and drug resistance. The NOTCH pathway, comprised of ligands JAG1, JAG2, DLL1, DLL3 and DLL4 and receptors NOTCH1, NOTCH2, NOTCH3 and NOTCH4, plays a critical role in stem cell signaling, cell proliferation, survival, apoptosis, and differentiation. Because of the existing body of data that implicates aberrant NOTCH pathway activation in resistance to chemotherapeutic agents in epithelial and hematologic tumors, we hypothesized that NOTCH inhibition would enhance the anti-tumor activity of chemotherapeutic agents. We have identified a NOTCH3 activating mutation in a patient breast cancer and developed a xenograft tumor model from the primary patient sample, OMP-B37. The combination of paclitaxel and OncoMed's anti-Notch2/3 antibody, OMP-59R5, inhibited the tumor growth of OMP-B37 xenograft tumors. In addition, we found that treatment with OMP-59R5 drove changes in tumor heterogeneity as indicated by the increased expression of markers associated with a mature cellular fate. The down-regulation of tumor growth promoting genes and NOTCH pathway markers was observed with OMP-59R5 treatment. The enhanced activity of combined OMP-59R5 and paclitaxel therapy in patient-derived tumor xenografts suggests that there is therapeutic potential in combining targeted NOTCH pathway inhibition with chemotherapeutic agents. Citation Format: Jie Wei, Jennifer Cain, Jalpa Shah, Breanna Wallace, Tracy Tang, Min Wang, Christopher Murriel, Cancilla Belinda, Austin Gurney, Aaron Sato, John Lewicki, Ann Kapoun, Tim Hoey. Combined therapy of a novel anti-NOTCH2/3 antibody with paclitaxel inhibits tumor growth in a patient-derived breast tumor xenograft. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Drug Sensitivity and Resistance: Improving Cancer Therapy; Jun 18-21, 2014; Orlando, FL. Philadelphia (PA): AACR; Clin Cancer Res 2015;21(4 Suppl): Abstract nr A33.
- Published
- 2015
- Full Text
- View/download PDF
48. A distinct strategy to generate high-affinity peptide binders to receptor tyrosine kinases
- Author
-
Q.L. Wu, Daniel J. Sexton, Daniel B. DeOliveira, Daniel T. Dransfield, Rolf E Swenson, E. Marinelli, Michael F. Tweedle, Greg Conley, M.A. von Wronski, Adrian D. Nunn, Q. Chang, Aaron K. Sato, A. Muruganandam, G. Connelly, Bo Song, Mary Devlin, Robert C. Ladner, Radhakrishna Pillai, P. Nanjappan, A. Shrivastava, A. Abujoub, C. Luneau, and Nancy J. Bogdan
- Subjects
VEGF receptors ,Bioengineering ,Peptide ,Biochemistry ,Chemical synthesis ,Receptor tyrosine kinase ,chemistry.chemical_compound ,Drug Delivery Systems ,stomatognathic system ,Competitive binding ,Peptide Library ,Protein Interaction Mapping ,Humans ,Molecular Biology ,chemistry.chemical_classification ,biology ,Receptor Protein-Tyrosine Kinases ,Drug Synergism ,Proto-Oncogene Proteins c-met ,Ligand (biochemistry) ,Vascular Endothelial Growth Factor Receptor-2 ,Monomer ,chemistry ,Drug Design ,biology.protein ,Peptides ,Dimerization ,Biotechnology ,Protein Binding - Abstract
We describe a novel and general way of generating high affinity peptide (HAP) binders to receptor tyrosine kinases (RTKs), using a multi-step process comprising phage-display selection, identification of peptide pairs suitable for hetero-dimerization (non-competitive and synergistic) and chemical synthesis of heterodimers. Using this strategy, we generated HAPs with K D s below 1 nM for VEGF receptor-2 (VEGFR-2) and c-Met. VEGFR-2 HAPs bound significantly better (6- to 500-fold) than either of the individual peptides that were used for heterodimer synthesis. Most significantly, HAPs were much better (150- to 800-fold) competitors than monomers of the natural ligand (VEGF) in various competitive binding and functional assays. In addition, we also found the binding of HAPs to be less sensitive to serum than their component peptides. We believe that this method may be applied to any protein for generating high affinity peptide (HAP) binders.
- Published
- 2005
49. Nitriles form mixed-coligand complexes with (99m)Tc-HYNIC-peptide
- Author
-
Ning Liu, Donald J. Hnatowich, Guozheng Liu, Aaron K. Sato, Yi Wang, Yu-Min Zhang, Charles R. Wescott, and Mary Rusckowski
- Subjects
Cancer Research ,Acetonitriles ,Nitrile ,Stereochemistry ,Ethylenediamine ,Peptide ,Conjugated system ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,Nitriles ,Animals ,Radiology, Nuclear Medicine and imaging ,Tissue Distribution ,Acetonitrile ,Chromatography, High Pressure Liquid ,chemistry.chemical_classification ,Tricine ,Ligand ,Nicotinic Acids ,Reversed-phase chromatography ,Organotechnetium Compounds ,Combinatorial chemistry ,Hydrazines ,chemistry ,Molecular Medicine - Abstract
Using a 12-amino acid peptide conjugated with HYNIC as a model, we investigated nitriles as possible coligands for labeling with 99m Tc. After the preparation of the 99m Tc labeled HYNIC-peptide using tricine as coligand, the addition of acetonitile was found by reverse phase HPLC to block further coligand exchange with ethylenediamine diacetic acid (EDDA) at room temperature. The addition of this nitrile changed the pharmacokinetics of the 99m Tc labeled peptide in normal mice towards faster clearance and significant differences in accumulation in most tissues sampled. By replacing acetonitrile with cyanoacetate, a nitrile not present in the HPLC eluant, it was possible to show the existence of a new, more hydrophilic, species by reverse phase HPLC. We conclude that nitriles can act as coligands for HYNIC-conjugated peptides labeled with 99m Tc and tricine. Furthermore, the presence of acetonitrile during Sep-Pak or HPLC purification may inadvertently generate a mixed tricine/acetonitile coligand 99m Tc-HYNIC-peptide complex.
- Published
- 2002
50. Abstract 213: Novel NOTCH3 activating mutations identified in tumors sensitive to OMP-59R5, a monoclonal antibody targeting the Notch2 and Notch3 receptors
- Author
-
Tracy Tang, Jalpa Shah, Jennifer Cain, Aaron K. Sato, Ann M. Kapoun, Belinda Cancilla, Breanna Wallace, Min Wang, Tim Hoey, Jie Wei, Austin L. Gurney, John Lewicki, and Chris Muriel
- Subjects
JAG2 ,Cancer Research ,Mutation ,JAG1 ,Mutant ,Notch signaling pathway ,Wild type ,Biology ,medicine.disease_cause ,Molecular biology ,Frameshift mutation ,Oncology ,Notch Family ,medicine - Abstract
The Notch signaling pathway, driven by four Notch family receptors (NOTCH1-4) and five canonical Notch ligands (DLL1, DLL3, DLL4, JAG1, and JAG2) in humans, critically regulates key functions during embryonic development as well as stem cell maintenance and differentiation in adult tissues. Altered Notch signaling activity has been documented in many types of cancers; depending on the tumor type, Notch signaling can be oncogenic or tumor suppressing. Activating mutations in NOTCH1 have been identified in hematopoietic cancers including T-cell acute lymphoblastic leukemia and chronic lymphoblastic leukemia. With the advance in high-throughput genomic sequencing platforms, mutations in the NOTCH genes have also been identified in solid tumors, albeit at much lower frequencies. Here we report for the first time the identification of two activating mutations in NOTCH3. One is a frameshift mutation in the PEST domain, and the other a frameshift mutation in the ankyrin (ANK) domain. The PEST frameshift mutation leads to the production of C-terminally truncated NOTCH3 protein, and by Western Blotting of nuclear fractions of tumor tissues and Immunohistochemistry (IHC) we show that this mutant NOTCH3 accumulates stably in the nuclei of the breast tumor in which it was identified. Breast xenograft tumors carrying this mutation are highly sensitive to OMP-59R5, a ligand-blocking antibody targeting the Notch2 and Notch3 receptors, suggesting that the mutation, although activating, is still ligand dependent. Consistent with this finding, expression of this mutant NOTCH3 constructed by site-directed mutagenesis exhibits higher baseline activity compared to wild type NOTCH3 and is equally responsive to ligands in in vitro NOTCH reporter assays. We are not able to observe increased nuclear accumulation of the ANK frameshift mutant NOTCH3 in the colon tumor in which it was identified. However, in vitro the ANK mutant NOTCH3 has higher baseline activity than wild-type NOTCH3 in the absence of ligands and remains highly responsive to ligands. The colon xenograft tumors carrying this NOTCH3 ANK mutation are also sensitive to OMP-59R5 in in vivo efficacy studies. Taken together our data suggest that NOTCH3 is oncogenic in certain subtypes of tumors, and tumors with these activating mutations may predictive sensitivity to therapeutic antibodies targeting the receptor. Citation Format: Breanna Wallace, Min Wang, Chris Muriel, Jennifer Cain, Belinda Cancilla, Jalpa Shah, Jie Wei, Austin Gurney, John Lewicki, Aaron Sato, Tim Hoey, Tracy Tang, Ann M. Kapoun. Novel NOTCH3 activating mutations identified in tumors sensitive to OMP-59R5, a monoclonal antibody targeting the Notch2 and Notch3 receptors. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 213. doi:10.1158/1538-7445.AM2013-213
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.