55 results on '"Manuel Baca"'
Search Results
2. 847 Pharmacokinetics and pharmacodynamics of GS-3583 in cynomolgus monkeys
- Author
-
Brian Carr, Michelle Kuhne, Hamlet Chu, Christopher Clarke, Manuel Baca, Magdeleine Hung, Mark Nagel, Alexandre Ambrogelly, and Nishanathan Rajakumaraswamy
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
3. A Therapeutic Uricase with Reduced Immunogenicity Risk and Improved Development Properties.
- Author
-
Andrew C Nyborg, Chris Ward, Anna Zacco, Benoy Chacko, Luba Grinberg, James C Geoghegan, Ryan Bean, Michaela Wendeler, Frank Bartnik, Ellen O'Connor, Flaviu Gruia, Vidyashankara Iyer, Hui Feng, Varnika Roy, Mark Berge, Jeffrey N Miner, David M Wilson, Dongmei Zhou, Simone Nicholson, Clynn Wilker, Chi Y Wu, Susan Wilson, Lutz Jermutus, Herren Wu, David A Owen, Jane Osbourn, Steven Coats, and Manuel Baca
- Subjects
Medicine ,Science - Abstract
Humans and higher primates are unique in that they lack uricase, the enzyme capable of oxidizing uric acid. As a consequence of this enzyme deficiency, humans have high serum uric acid levels. In some people, uric acid levels rise above the solubility limit resulting in crystallization in joints, acute inflammation in response to those crystals causes severe pain; a condition known as gout. Treatment for severe gout includes injection of non-human uricase to reduce serum uric acid levels. Krystexxa® is a hyper-PEGylated pig-baboon chimeric uricase indicated for chronic refractory gout that induces an immunogenic response in 91% of treated patients, including infusion reactions (26%) and anaphylaxis (6.5%). These properties limit its use and effectiveness. An innovative approach has been used to develop a therapeutic uricase with improved properties such as: soluble expression, neutral pH solubility, high E. coli expression level, thermal stability, and excellent activity. More than 200 diverse uricase sequences were aligned to guide protein engineering and reduce putative sequence liabilities. A single uricase lead candidate was identified, which showed low potential for immunogenicity in >200 human donor samples selected to represent diverse HLA haplotypes. Cysteines were engineered into the lead sequence for site specific PEGylation and studies demonstrated >95% PEGylation efficiency. PEGylated uricase retains enzymatic activity in vitro at neutral pH, in human serum and in vivo (rats and canines) and has an extended half-life. In canines, an 85% reduction in serum uric acid levels was observed with a single subcutaneous injection. This PEGylated, non-immunogenic uricase has the potential to provide meaningful benefits to patients with gout.
- Published
- 2016
- Full Text
- View/download PDF
4. Supplementary Figure 2 from Multivalent Scaffold Proteins as Superagonists of TRAIL Receptor 2–Induced Apoptosis
- Author
-
Manuel Baca, David A. Tice, Herren Wu, Ching Ching Leow, Ivan Inigo, Zhan Xiao, Rosa Carrasco, Kristen Lekstrom, Hui Feng, Lin Wang, Luba Grinberg, and Jeffery S. Swers
- Abstract
PDF file - 85K, Supplementary Figure S2: Specificity of TRAILR2-induced cell killing. TRAIL-sensitive cell lines were incubated with 100pM G6T8 and different concentrations of soluble TRAILR2-Fc fusion protein.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Methods from Multivalent Scaffold Proteins as Superagonists of TRAIL Receptor 2–Induced Apoptosis
- Author
-
Manuel Baca, David A. Tice, Herren Wu, Ching Ching Leow, Ivan Inigo, Zhan Xiao, Rosa Carrasco, Kristen Lekstrom, Hui Feng, Lin Wang, Luba Grinberg, and Jeffery S. Swers
- Abstract
PDF file - 95K
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 4 from Multivalent Scaffold Proteins as Superagonists of TRAIL Receptor 2–Induced Apoptosis
- Author
-
Manuel Baca, David A. Tice, Herren Wu, Ching Ching Leow, Ivan Inigo, Zhan Xiao, Rosa Carrasco, Kristen Lekstrom, Hui Feng, Lin Wang, Luba Grinberg, and Jeffery S. Swers
- Abstract
PDF file - 98K, Supplementary Figure S4: Caspase activation by G6T8.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 3 from Multivalent Scaffold Proteins as Superagonists of TRAIL Receptor 2–Induced Apoptosis
- Author
-
Manuel Baca, David A. Tice, Herren Wu, Ching Ching Leow, Ivan Inigo, Zhan Xiao, Rosa Carrasco, Kristen Lekstrom, Hui Feng, Lin Wang, Luba Grinberg, and Jeffery S. Swers
- Abstract
PDF file - 59K, Supplementary Figure S3: G6T8 is specific for human TRAILR2 and does not cross-react with other TRAIL receptors.
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Multivalent Scaffold Proteins as Superagonists of TRAIL Receptor 2–Induced Apoptosis
- Author
-
Manuel Baca, David A. Tice, Herren Wu, Ching Ching Leow, Ivan Inigo, Zhan Xiao, Rosa Carrasco, Kristen Lekstrom, Hui Feng, Lin Wang, Luba Grinberg, and Jeffery S. Swers
- Abstract
Activation of TNF-related apoptosis-inducing ligand receptor 2 (TRAILR2) can induce apoptosis in a variety of human cancer cell lines and xenografts, while lacking toxicity in normal cells. The natural ligand and agonistic antibodies show antitumor activity in preclinical models of cancer, and this had led to significant excitement in the clinical potential of these agents. Unfortunately, this optimism has been tempered by trial data that, thus far, are not showing clear signs of efficacy in cancer patients. The reasons for discrepant preclinical and clinical observations are not understood, but one possibility is that the current TRAILR2 agonists lack sufficient potency to achieve a meaningful response in patients. Toward addressing that possibility, we have developed multivalent forms of a new binding scaffold (Tn3) that are superagonists of TRAILR2 and can induce apoptosis in tumor cell lines at subpicomolar concentrations. The monomer Tn3 unit was a fibronectin type III domain engineered for high-affinity TRAILR2 binding. Multivalent presentation of this basic unit induced cell death in TRAILR2-expressing cell lines. Optimization of binding affinity, molecular format, and valency contributed to cumulative enhancements of agonistic activity. An optimized multivalent agonist consisting of 8 tandem Tn3 repeats was highly potent in triggering cell death in TRAIL-sensitive cell lines and was 1 to 2 orders of magnitude more potent than TRAIL. Enhanced potency was also observed in vivo in a tumor xenograft setting. The TRAILR2 superagonists described here have the potential for superior clinical activity in settings insensitive to the current therapeutic agonists that target this pathway. Mol Cancer Ther; 12(7); 1235–44. ©2013 AACR.
- Published
- 2023
- Full Text
- View/download PDF
9. Impact of maternal diphtheria-tetanus-acellular pertussis vaccination on pertussis booster immune responses in toddlers: Follow-up of a randomized trial
- Author
-
Esperanza Escribano Palomino, Federico Martinón-Torres, Kirsten P Perrett, Manuel Baca, Mariano Miranda-Valdivieso, Jan Janota, Brigitte Cheuvart, Scott A. Halperin, Otto G. Vanderkooi, Stranák Z, Miia Virta, Terry Nolan, Paola Marchisio, Sarka Rumlarova, Lusine Kostanyan, Narcisa Mesaros, Sherine Kuriyakose, José Garcia-Sicilia, Begoña Arias Novas, María José Cilleruelo Ortega, Ignacio Salamanca de la Cueva, Pavel Kosina, Maria Angeles Ceregido, Jose Manuel Merino Arribas, José Tomás Ramos Amador, Gian Vincenzo Zuccotti, Jan Bozensky, Nadia Meyer, Bruce Tapiero, Tampere University, and Clinical Medicine
- Subjects
Whooping Cough ,Pneumococcal conjugate vaccine ,0302 clinical medicine ,Pregnancy ,030212 general & internal medicine ,Haemophilus Vaccines ,Tetanus ,Vaccination ,Toxoid ,Diphtheria ,Antibodies, Bacterial ,Europe ,Blunting ,Infectious Diseases ,Child, Preschool ,Molecular Medicine ,Female ,Pertactin ,medicine.drug ,Canada ,Immunization, Secondary ,Diphtheria-Tetanus-acellular Pertussis Vaccines ,complex mixtures ,03 medical and health sciences ,Pertussis ,030225 pediatrics ,medicine ,Humans ,Vaccines, Combined ,Diphtheria-Tetanus-Pertussis Vaccine ,Toddlers ,Reactogenicity ,General Veterinary ,General Immunology and Microbiology ,business.industry ,Australia ,Immunity ,Public Health, Environmental and Occupational Health ,Infant ,medicine.disease ,Booster ,Poliovirus Vaccine, Inactivated ,Immunization ,Maternal immunization ,Immunology ,3111 Biomedicine ,business ,Tdap vaccine ,Follow-Up Studies - Abstract
Background: Transplacentally transferred antibodies induced by maternal pertussis vaccination interfere with infant immune responses to pertussis primary vaccination. We evaluated whether this interference remains in toddlers after booster vaccination. Methods: In a prior phase IV, observer-blind, placebo-controlled, randomized study (NCT02377349), pregnant women in Australia, Canada and Europe received intramuscular tetanus-reduced-antigen-content diphtheria-three-component acellular pertussis vaccine (Tdap group) or placebo (control group) at 270/7–366/7 weeks’ gestation, with crossover immunization postpartum. Their infants were primed (study NCT02422264) and boosted (at 11–18 months; current study NCT02853929) with diphtheria-tetanus-three-component acellular pertussis-hepatitis B virus-inactivated poliovirus/Haemophilus influenzae type b vaccine (DTaP-HepB-IPV/Hib) and 13-valent pneumococcal conjugate vaccine. Immunogenicity before and after booster vaccination, and reactogenicity and safety of the booster were evaluated descriptively. Results: 263 (Tdap group) and 277 (control group) toddlers received a DTaP-HepB-IPV/Hib booster. Pre-booster vaccination, observed geometric mean concentrations (GMCs) for the three pertussis antigens and diphtheria were 1.4–1.5-fold higher in controls than in the Tdap group. No differences were observed for the other DTaP-HepB-IPV/Hib antigens. One month post-booster vaccination, booster response rates for pertussis antigens were ≥ 92.1% and seroprotection rates for the other DTaP-HepB-IPV/Hib antigens were ≥ 99.2% in both groups (primary objective). Higher post-booster GMCs were observed in controls versus the Tdap group for anti-filamentous hemagglutinin (1.2-fold), anti-pertussis toxoid (1.5-fold) and anti-diphtheria (1.4-fold). GMCs for the other DTaP-HepB-IPV/Hib antigens were similar between groups. Serious adverse events were reported for three toddlers (controls, not vaccination-related). One death occurred pre-booster (Tdap group, not vaccination-related). Conclusions: As a consequence of interference of maternal pertussis antibodies with infant immune responses to pertussis primary vaccination, pertussis antibody concentrations were still lower in toddlers from Tdap-vaccinated mothers before DTaP-HepB-IPV/Hib booster vaccination. After the booster, antibody concentrations were lower for filamentous hemagglutinin and pertussis toxoid but not for pertactin. The clinical significance of this interference requires further evaluation. Clinical Trial Registration. ClinicalTrials.gov: NCT02853929. publishedVersion
- Published
- 2021
- Full Text
- View/download PDF
10. Group 1 ILCs regulate T cell–mediated liver immunopathology by controlling local IL-2 availability
- Author
-
Valeria Fumagalli, Valentina Venzin, Pietro Di Lucia, Federica Moalli, Xenia Ficht, Gioia Ambrosi, Leonardo Giustini, Francesco Andreata, Marta Grillo, Diletta Magini, Micol Ravà, Christin Friedrich, Jason D. Fontenot, Philippe Bousso, Sarah A. Gilmore, Shahzada Khan, Manuel Baca, Eric Vivier, Georg Gasteiger, Mirela Kuka, Luca G. Guidotti, Matteo Iannacone, IRCCS San Raffaele Scientific Institute [Milan, Italie], Universita Vita Salute San Raffaele = Vita-Salute San Raffaele University [Milan, Italie] (UniSR), Max Planck Research Group - The Julius-Maximiliams-Universität Würzburg, Sangamo Therapeutics brisbane, Dynamiques des Réponses immunes - Dynamics of Immune Responses, Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Gilead Sciences, Centre d'Immunologie de Marseille - Luminy (CIML), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Innate Pharma, Hôpital de la Timone [CHU - APHM] (TIMONE), ANR-17-RHUS-0007,PIONEER,Precision Immuno-Oncology for advanced Non small cell lung cancer patients with PD-1 ICI Resistance(2017), Fumagalli, V, Venzin, V, Di Lucia, P, Moalli, F, Ficht, X, Ambrosi, G, Giustini, L, Andreata, F, Grillo, M, Magini, D, Rava, M, Friedrich, C, Fontenot, J, Bousso, P, Gilmore, S, Khan, S, Baca, M, Vivier, E, Gasteiger, G, Kuka, M, Guidotti, L, Iannacone, M, Fumagalli, V., Venzin, V., Di Lucia, P., Moalli, F., Ficht, X., Ambrosi, G., Giustini, L., Andreata, F., Grillo, M., Magini, D., Rava, M., Friedrich, C., Fontenot, J. D., Bousso, P., Gilmore, S. A., Khan, S., Baca, M., Vivier, E., Gasteiger, G., Kuka, M., Guidotti, L. G., and Iannacone, M.
- Subjects
Mice, Inbred BALB C ,Interleukin-2 (IL-2) ,[SDV]Life Sciences [q-bio] ,Immunology ,Mice, Transgenic ,General Medicine ,CD8-Positive T-Lymphocytes ,Immunity, Innate ,Hepatitis B Virus (HBV) ,Killer Cells, Natural ,Mice, Inbred C57BL ,Mice ,Mice, Congenic ,Group 1 innate lymphoid cells (ILCs) ,Animals ,Interleukin-2 ,Lymphocytes - Abstract
Group 1 innate lymphoid cells (ILCs), which comprise both natural killer (NK) cells and ILC1s, are important innate effectors that can also positively and negatively influence adaptive immune responses. The latter function is generally ascribed to the ability of NK cells to recognize and kill activated T cells. Here, we used multiphoton intravital microscopy in mouse models of hepatitis B to study the intrahepatic behavior of group 1 ILCs and their cross-talk with hepatitis B virus (HBV)–specific CD8+T cells. We found that hepatocellular antigen recognition by effector CD8+T cells triggered a prominent increase in the number of hepatic NK cells and ILC1s. Group 1 ILCs colocalized and engaged in prolonged interactions with effector CD8+T cells undergoing hepatocellular antigen recognition; however, they did not induce T cell apoptosis. Rather, group 1 ILCs constrained CD8+T cell proliferation by controlling local interleukin-2 (IL-2) availability. Accordingly, group 1 ILC depletion, or genetic removal of their IL-2 receptor a chain, considerably increased the number of intrahepatic HBV-specific effector CD8+T cells and the attendant immunopathology. Together, these results reveal a role for group 1 ILCs in controlling T cell–mediated liver immunopathology by limiting local IL-2 concentration and have implications for the treatment of chronic HBV infection.
- Published
- 2022
- Full Text
- View/download PDF
11. LB13. The Efficacy and Impact in Heathy Infants of Nirsevimab on Medically Attended RSV Lower Respiratory Tract Infection
- Author
-
Hammitt, Laura, primary, Hammitt, Laura, additional, Dagan, Ron, additional, Yuan, Yuan, additional, Cots, Manuel Baca, additional, Bosheva, Miroslava, additional, Mahdi, Shabhir A, additional, Muller, William J, additional, Zar, Heather J, additional, Brooks, Dennis, additional, Grenham, Amy, additional, Hamrén, Ulrika Wählby, additional, Mankad, Vaishali S, additional, Ren, Pin, additional, Takas, Therese, additional, Heinrichs, Jon, additional, Leach, Amanda, additional, Griffin, M Pamela, additional, and Villafana, Tonya L, additional
- Published
- 2021
- Full Text
- View/download PDF
12. LB13. The Efficacy and Impact in Heathy Infants of Nirsevimab on Medically Attended RSV Lower Respiratory Tract Infection
- Author
-
Laura Hammitt, Ron Dagan, Yuan Yuan, Manuel Baca Cots, Miroslava Bosheva, Shabhir A Mahdi, William J Muller, Heather J Zar, Dennis Brooks, Amy Grenham, Ulrika Wählby Hamrén, Vaishali S Mankad, Pin Ren, Therese Takas, Jon Heinrichs, Amanda Leach, M Pamela Griffin, and Tonya L Villafana
- Subjects
Infectious Diseases ,AcademicSubjects/MED00290 ,Oncology ,Late Breaker Abstracts - Abstract
Background Respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract infection (LRTI) in infants. Nirsevimab is a single-dose monoclonal antibody with extended half-life that was shown to protect preterm infants 29 to < 35 weeks gestation against RSV LRTI. However, most medically attended (MA) cases occur in otherwise healthy, term infants for whom there is currently no effective RSV prevention strategy. We report the primary analysis of efficacy and safety, along with the impact of nirsevimab in late preterm and term infants (≥ 35 weeks gestation) in the phase 3 MELODY study (NCT03979313). Methods Infants were randomized 2:1 to receive one intramuscular injection of nirsevimab (50 mg if < 5 kg; 100 mg if ≥ 5 kg at dosing) or placebo entering their first RSV season. The primary endpoint was the incidence of MA RSV LRTI over 150 days postdose. Cases met predefined clinical criteria of disease severity and were confirmed by real-time reverse-transcriptase PCR. Safety was evaluated through 360 days postdose. Enrollment started on 23 July 2019 and was suspended following the declaration of the COVID-19 pandemic by the WHO on 11 March 2020. Results Overall, 1490 infants were randomized and included in the intent-to-treat population; 1465 (98%) completed the 150-day efficacy follow-up, and 1367 (92%) completed the 360-day safety follow-up. The incidence of MA RSV LRTI was 1.2% (n=12/994) in the nirsevimab group and 5.0% (n=25/496) in the placebo group, giving nirsevimab an efficacy of 74.5% (95% confidence interval [CI]: 49.6, 87.1; p< 0.0001). Nirsevimab averted 93.6 (95% CI 63.0, 124.0) MA LRTIs per 1000 infants dosed. Nirsevimab was well tolerated, with similar rates of adverse events (87.4% nirsevimab; 86.8% placebo) and serious adverse events (6.8% nirsevimab; 7.3% placebo) between groups. Conclusion In this phase 3 study, a single dose of nirsevimab protected late preterm and term infants against MA RSV LRTI over an RSV season with a favorable safety profile. Approximately 11 infants need to be immunized to prevent 1 case of LRTI; nirsevimab has the potential to be an important intervention to reduce the burden of RSV LRTI in healthy infants. Disclosures Laura Hammitt, MD, MedImmune (Grant/Research Support, Scientific Research Study Investigator, Research Grant or Support)Merck & Co., Inc. (Grant/Research Support, Scientific Research Study Investigator, Research Grant or Support)Novavax (Grant/Research Support, Scientific Research Study Investigator, Research Grant or Support)Pfizer (Grant/Research Support, Scientific Research Study Investigator, Research Grant or Support) Laura Hammitt, MD, MedImmune (Individual(s) Involved: Self): Grant/Research Support, Research grant to my institution; Merck (Individual(s) Involved: Self): Grant/Research Support, Research grant to my institution; Pfizer (Individual(s) Involved: Self): Grant/Research Support, Research grant to my institution Ron Dagan, MD, Medimmune/AstraZeneca (Grant/Research Support, Scientific Research Study Investigator, Research Grant or Support)MSD (Consultant, Grant/Research Support, Scientific Research Study Investigator, Advisor or Review Panel member, Research Grant or Support, Speaker’s Bureau)Pfizer (Consultant, Grant/Research Support, Scientific Research Study Investigator, Advisor or Review Panel member, Research Grant or Support, Speaker’s Bureau) Yuan Yuan, PhD, AstraZeneca (Employee, Shareholder) Shabhir A. Mahdi, PhD, BMGF (Research Grant or Support)EDCTP (Research Grant or Support)GlaxoSmithKline (Research Grant or Support)Melody (Research Grant or Support)Minervax (Research Grant or Support)Novavax (Research Grant or Support)SAMRC (Research Grant or Support) William J. Muller, MD, PhD, Ansun (Scientific Research Study Investigator)Astellas (Scientific Research Study Investigator)AstraZeneca (Scientific Research Study Investigator)Genentech (Scientific Research Study Investigator)Gilead (Scientific Research Study Investigator)Janssen (Scientific Research Study Investigator)Karius (Scientific Research Study Investigator)Melinta (Scientific Research Study Investigator)Merck (Scientific Research Study Investigator)Nabriva (Scientific Research Study Investigator)Seqirus (Scientific Research Study Investigator)Tetraphase (Scientific Research Study Investigator) William J. Muller, MD, PhD, Ansun (Individual(s) Involved: Self): Grant/Research Support; Astellas (Individual(s) Involved: Self): Research Grant or Support; AstraZeneca (Individual(s) Involved: Self): Grant/Research Support; BD (Individual(s) Involved: Self): Research Grant or Support; Eli Lilly (Individual(s) Involved: Self): Grant/Research Support; Gilead (Individual(s) Involved: Self): Grant/Research Support; Karius, Inc. (Individual(s) Involved: Self): Grant/Research Support, Scientific Research Study Investigator; Melinta (Individual(s) Involved: Self): Grant/Research Support; Merck (Individual(s) Involved: Self): Grant/Research Support; Moderna (Individual(s) Involved: Self): Grant/Research Support; Nabriva (Individual(s) Involved: Self): Grant/Research Support; Seqirus (Individual(s) Involved: Self): Consultant; Tetraphase (Individual(s) Involved: Self): Grant/Research Support Heather J. Zar, PhD, AstraZeneca (Grant/Research Support)Novavax (Grant/Research Support)Pfizer (Grant/Research Support, Advisor or Review Panel member) Dennis Brooks, MD, AstraZeneca (Employee) Amy Grenham, MSc, AstraZeneca (Employee, Shareholder) Ulrika Wählby Hamrén, PhD, AstraZeneca R&D (Employee, Shareholder) Vaishali S. Mankad, MD, AstraZeneca (Employee) Therese Takas, BSc, AstraZeneca (Employee, Other Financial or Material Support, Own stock in AstraZeneca) Jon Heinrichs, PhD, AstraZeneca (Shareholder)Bristol Myers Squibb (Shareholder)J&J (Shareholder)Merck (Shareholder)Organon (Shareholder)Procter & Gamble (Shareholder)Sanofi (Shareholder)Sanofi Pasteur (Employee) Amanda Leach, MRCPCH, AstraZeneca (Employee, Shareholder) M. Pamela Griffin, MD, AstraZeneca (Employee) Tonya L. Villafana, PhD, AstraZeneca (Employee)
- Published
- 2021
13. Immunantwort auf die DTPa-HBV-IPV/Hib-Auffrischimpfung bei Kleinkindern von Müttern, die während der Schwangerschaft mit Tdap-Impfstoff geimpft worden waren: Folgestudie einer randomisierten, placebokontrollierten Studie
- Author
-
Terry Nolan, P Kosina, Federico Martinón-Torres, Paola Marchisio, Mariano Miranda-Valdivieso, Miia Virta, Lusine Kostanyan, José Garcia-Sicilia, Brigitte Cheuvart, Kirsten P Perrett, M.J Cilleruelo Ortega, Sherine Kuriyakose, Narcisa Mesaros, Manuel Baca, Scott A. Halperin, JT Ramos Amador, Maria Angeles Ceregido, IS de la Cueva, Otto G. Vanderkooi, B Arias Novas, Jan Janota, J Bozensky, Palomino E Escribano, Bruce Tapiero, Gian Vincenzo Zuccotti, Nadia Meyer, Z Stranak, and JM Merino Arribas
- Published
- 2020
- Full Text
- View/download PDF
14. 846 Pre-clinical validation of a FLT3L-fusion protein for dendritic cell expansion and anti-tumor efficacy
- Author
-
Brian I. Carr, Hamlet Chu, Mark Nagel, Nicholas J. Wilson, Magdeleine Hung, Sarah Ng, Michelle R. Kuhne, Alexandre Ambrogelly, Christopher Clarke, and Manuel Baca
- Subjects
Pharmacology ,Antitumor activity ,Cancer Research ,Oncology ,Chemistry ,Immunology ,Cancer research ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Molecular Medicine ,Immunology and Allergy ,Dendritic cell ,Fusion protein ,RC254-282 - Abstract
BackgroundThe ligand for the receptor tyrosine kinase FMS-like tyrosine kinase 3 (FLT3) plays an importantrole in hematopoiesis. FLT3 signaling is required for the differentiation andexpansion of dendritic cells. In the context of cancer immunity, the conventional dendritic cellsubtype 1 (cDC1) are required for the generation of tumor-specific T cell responses in mousepreclinical models. In human tumors cDC1 are often underrepresented in thetumor microenvironment, supporting the hypothesis that therapeutically increasing their number via FLT3 pathway stimulation has the potential to promote T cell-mediated anti-tumor efficacy.MethodsGS-3583 is a fusion protein composed of the extracellular domain (ECD) of human FLT3 ligand(FLT3L) combined with a modified fragment crystallizable (Fc) region of human IgG4. GS-3583was designed to induce cDC1 expansion and subsequently promote tumor-reactive T cell priming, activation and recruitment into the tumor microenvironment. To evaluate the therapeutic efficacy of FLT3 stimulation in vivo, a mouse surrogate mGS-3583was designed using the ECD of mouse FLT3L fused to an engineered mouse IgG2a Fc withattenuated binding to mouse FcgRs.Results mGS-3583 bound to recombinant mouse FLT3 with an estimated affinity of 15 nM, and to mouse FLT3-expressing cells with an EC50 of 0.15 nM. In vivo, mGS-3583 showed single agent dose-dependent tumor growth inhibition (TGI) in tumors that correlated with peripheral and intratumoral cDC1 expansion. In tumors with no initial immune infiltration, mGS-3583 led to an influx of T cells into the tumors. In addition to single agent efficacy, mGS-3583 combined effectively with programmed cell death protein (ligand)-1 (PD(L)-1) pathway blockade.ConclusionsIn vivo expansion of dendritic cells can convert uninflamed (cold) tumors to immunologically active (hot) tumors and initiate productive anti-tumor immune responses. These findings support the development GS-3583 as a promising candidate for cancer immunotherapy.
- Published
- 2021
- Full Text
- View/download PDF
15. 847 Pharmacokinetics and pharmacodynamics of GS-3583 in cynomolgus monkeys
- Author
-
Mark Nagel, Michelle R. Kuhne, Manuel Baca, Christopher Clarke, Magdeleine Hung, Nishanathan Rajakumaraswamy, Alexandre Ambrogelly, Hamlet Chu, and Brian I. Carr
- Subjects
Pharmacology ,Cancer Research ,Oncology ,Pharmacokinetics ,business.industry ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business ,RC254-282 - Abstract
BackgroundThe ligand for the receptor tyrosine kinase FMS-like tyrosine kinase 3 (FLT3) plays an importantrole in hematopoiesis. FLT3 signaling is required for the differentiation andexpansion of dendritic cells. In the context of cancer immunity, the conventional dendritic cellsubtype 1 (cDC1) are required for the generation of tumor-specific T cell responses in mousepreclinical models. In human tumors cDC1 are often underrepresented in thetumor microenvironment, supporting the hypothesis that therapeutically increasing their number via FLT3 pathway stimulation has the potential to promote T cell-mediated anti-tumor activity.MethodsGS-3583 is a fusion protein composed of the extracellular domain of human FLT3 ligand(FLT3L) combined with a modified fragment crystallizable (Fc) region of human IgG4. GS-3583was designed to induce cDC1 expansion and subsequently promote tumor-reactive T cell priming, activation and recruitment into the tumor microenvironment. The pharmacokinetics (PK) and pharmacodynamics (PD) of GS-3583 has been characterized in a 4-week repeat dose GLP study in cynomolgus monkeys at doses ranging from 0.3 to 10mg/kg GS-3583 was given as an intravenous injection.ResultsImmunophenotyping analysis of peripheral blood cells from GS-3583 treated monkeys demonstrated a non-dose-dependent expansion of cDC1 and cDC2 populations. The peak expansion for cDC1 and cDC2 occurred at Day 8 to Day 15. At peak, there was a 160-fold relative increase in cDC1 and 150-fold increase in cDC2 at the highest dose tested. There were dose-dependent increases in the exposure (AUC and Cmax) of GS-3583. GS-3583 was well-tolerated with no mortality or adverse clinical signs.ConclusionsThe administration of GS-3583 leads to increases in cDC1 and cDC2 populations. It was well tolerated at the maximal dose tested with no adverse clinical signs. Further clinical development of GS-3583 is warranted.
- Published
- 2021
- Full Text
- View/download PDF
16. Impact of tetanus-diphtheria-acellular pertussis immunization during pregnancy on subsequent infant immunization seroresponses: follow-up from a large randomized placebo-controlled trial
- Author
-
Terry Nolan, Brigitte Cheuvart, Felix Omeñaca, Lusine Kostanyan, Narcisa Mesaros, Paola Marchisio, Federico Martinón-Torres, Alfonso Carmona Martinez, Mariano Miranda-Valdivieso, Jose Manuel Merino Arribas, José Garcia-Sicilia, José Tomás Ramos Amador, Nadia Meyer, Kirsten P Perrett, Scott A. Halperin, Gian Vincenzo Zuccotti, Otto G. Vanderkooi, Sherine Kuriyakose, Begoña Arias Novas, Z Stranak, María José Cilleruelo Ortega, Manuel Baca, Paolo Manzoni, Maria Angeles Ceregido, and Miia Virta
- Subjects
Pediatrics ,Pneumococcal conjugate vaccine ,Pneumococcal Vaccines ,0302 clinical medicine ,Pregnancy ,Medicine ,030212 general & internal medicine ,Haemophilus Vaccines ,Vaccines ,Tetanus ,Combined ,Bacterial ,Antibodies, Bacterial ,Vaccination ,Poliovirus Vaccine ,Blunting ,Infectious Diseases ,Infants ,Maternal immunization ,Pertussis ,Tdap vaccine ,Diphtheria-Tetanus-Pertussis Vaccine ,Diphtheria-Tetanus-acellular Pertussis Vaccines ,Female ,Follow-Up Studies ,Hepatitis B Vaccines ,Humans ,Infant ,Poliovirus Vaccine, Inactivated ,Vaccines, Combined ,Molecular Medicine ,medicine.drug ,medicine.medical_specialty ,complex mixtures ,Antibodies ,03 medical and health sciences ,030225 pediatrics ,Immunization during pregnancy ,Reactogenicity ,General Veterinary ,General Immunology and Microbiology ,business.industry ,Diphtheria ,Public Health, Environmental and Occupational Health ,Inactivated ,medicine.disease ,Immunization ,business - Abstract
Background Pertussis immunization during pregnancy results in high pertussis antibody concentrations in young infants but may interfere with infant immune responses to post-natal immunization. Methods This phase IV, multi-country, open-label study assessed the immunogenicity and safety of infant primary vaccination with DTaP-HepB-IPV/Hib and 13-valent pneumococcal conjugate vaccine (PCV13). Enrolled infants (6–14 weeks old) were born to mothers who were randomized to receive reduced-antigen-content diphtheria-tetanus-three-component acellular pertussis vaccine (Tdap group) or placebo (control group) during pregnancy (270/7–366/7 weeks’ gestation) with crossover immunization postpartum. All infants received 2 or 3 DTaP-HepB-IPV/Hib and PCV13 doses according to national schedules. Immunogenicity was assessed in infants pre- and 1 month post-primary vaccination. The primary objective was to assess seroprotection/vaccine response rates for DTaP-HepB-IPV/Hib antigens 1 month post-primary vaccination. Results 601 infants (Tdap group: 296; control group: 305) were vaccinated. One month post-priming, seroprotection rates were 100% (diphtheria; tetanus), ≥98.5% (hepatitis B), ≥95.9% (polio) and ≥94.5% (Hib) in both groups. Vaccine response rates for pertussis antigens were significantly lower in infants whose mothers received pregnancy Tdap (37.5–77.1%) versus placebo (90.0–99.2%). Solicited and unsolicited adverse event rates were similar between groups. Serious adverse events occurred in 2.4% (Tdap group) and 5.6% (control group) of infants, none were vaccination-related. Conclusions Pertussis antibodies transferred during pregnancy may decrease the risk of pertussis infection in the first months of life but interfere with the infant’s ability to produce pertussis antibodies, the clinical significance of which remains unknown. Safety and reactogenicity results were consistent with previous experience. Clinical Trial Registration: ClinicalTrials.gov: NCT02422264.
- Published
- 2019
17. Efficacy, immunogenicity, and safety of a quadrivalent inactivated influenza vaccine in children aged 6-35 months: A multi-season randomised placebo-controlled trial in the Northern and Southern Hemispheres
- Author
-
Pepin, Stephanie Dupuy, Martin Corazon Borja-Tabora, Charissa Fay Montellano, May Bravo, Lulu Santos, Jaime de Castro, Jo-Anne Rivera-Medina, Doris Maribel Cutland, Clare Ariza, Miguel Diez-Domingo, Javier Diaz Gonzalez, Celia and Martinon-Torres, Federico Papadopoulou-Alataki, Efimia and Theodoriado, Maria Kazek-Duret, Marie Pierre Gurunathan, Sanjay and De Bruijn, Iris Abalos, Karina Aurell, Helena Maria Baldo, Jose Bona, Gianni Angel, Miguel Cadorna-Carlos, Josefina Cangrejo, Marcela Capilna, Brindusa Ruxandra Cara, Alexandra Carmen Carmona Martinez, Alfonso Chemin, Frederic and Closa, Ricardo Cots, Manuel Baca Coux, Florence and Diez-Domingo, Javier Dracea, Laura Larisa Emporiadou, Maria and Espiau, Maria Esposito, Susanna Pecurariu, Oana Asso Falup and Garces-Sanchez, Maria Garg, Sanjay Gil, Amparo Gonzales, Laurie Guevel, Ronan Guillen, Sara Icardi, Giancarlo and Laot, Thelma Lacroix, Isabelle Mares, Josep Martinez Pons, Manuel Moreau, Catherine Neamtu, Mihai Leonida Neculau, Andrea Elena Ojeda, Joyce Papaevangelou, Vana Penon, Maria Gabriella Petit, Celine Philibert, Marie Planelles Cantarino, Victoria Py, Marie-Laure Ramos, Jose T. Rivas, Enrique Roilides, Emmanouel Rosich, Angels Salamand, Camille and Suarez, Eva Surdu, Gabriel Doru Cerdan Vera, Teresa and Tsolia, Mariza Vicedo, Mira Woods, Anne GQM05 Study Grp
- Abstract
Background: A quadrivalent split-virion inactivated influenza vaccine (VaxigripTetre (TM), Sanofi Pasteur; IIV4) containing two A strains (H1N1 and H3N2) and B strains from both lineages (Victoria and Yamagata) was approved in Europe in 2016 for individuals aged >= 3 years. This study examined the efficacy and safety of IIV4 in children aged 6-35 months. Methods: This was a phase III randomised controlled trial conducted in Latin America, Asia, Africa, and Europe during the Northern Hemisphere 2014/2015 and 2015/2016 and Southern Hemisphere 2014 and 2015 influenza seasons. Healthy children aged 6-35 months not previously vaccinated against influenza were randomised to receive two full doses 28 days apart of IIV4, placebo, the licensed trivalent splitvirion inactivated vaccine (IIV3), an investigational IIV3 containing a B strain from the alternate lineage. The primary objective was to demonstrate efficacy against influenza illness caused by any strain or vaccine-similar strains. Results: The study enrolled 5806 participants. Efficacy, assessed in 4980 participants completing the study according to protocol, was demonstrated for IIV4. Vaccine efficacy was 50.98% (97% CI, 37.36-61.86%) against influenza caused by any A or B type and 68.40% (97% CI, 47.07-81.92%) against influenza caused by vaccine-like strains. Safety profiles were similar for IIV4, placebo, and the IIV3s, although injection-site reactions were slightly more frequent for IIV4 than placebo. Conclusions: IIV4 was safe and effective for protecting children aged 6-35 months against influenza illness caused by vaccine-similar or any circulating strains. (C) 2018 The Authors. Published by Elsevier Ltd.
- Published
- 2019
18. Crystal structure of the human 4-1BB/4-1BBL complex
- Author
-
Ryan N. Gilbreth, Luba Grinberg, Vaheh Oganesyan, Hamza Amdouni, Shabazz Novarra, Arnita Barnes, and Manuel Baca
- Subjects
0301 basic medicine ,Protein Conformation ,Trimer ,Crystallography, X-Ray ,Ligands ,Biochemistry ,Protein–protein interaction ,03 medical and health sciences ,Tumor Necrosis Factor Receptor Superfamily, Member 9 ,0302 clinical medicine ,Protein structure ,Humans ,Receptor ,Molecular Biology ,Binding Sites ,Chemistry ,Cell Biology ,Ligand (biochemistry) ,Cell biology ,030104 developmental biology ,4-1BB Ligand ,HEK293 Cells ,Structural biology ,030220 oncology & carcinogenesis ,Protein Structure and Folding ,Tumor necrosis factor alpha ,Protein Multimerization ,Function (biology) ,Protein Binding - Abstract
4-1BBL is a member of the tumor necrosis factor (TNF) superfamily and is the ligand for the TNFR superfamily receptor, 4-1BB. 4-1BB plays an immunomodulatory role in T cells and NK cells, and agonists of this receptor have garnered strong attention as potential immunotherapy agents. Broadly speaking, the structural features of TNF superfamily members, their receptors, and ligand-receptor complexes are similar. However, a published crystal structure of human 4-1BBL suggests that it may be unique in this regard, exhibiting a three-bladed propeller-like trimer assembly that is distinctly different from that observed in other family members. This unusual structure also suggests that the human 4-1BB/4-1BBL complex may be structurally unique within the TNF/TNFR superfamily, but to date no structural data have been reported. Here we report the crystal structure of the human 4-1BB/4-1BBL complex at 2.4-A resolution. In this structure, 4-1BBL does not adopt the unusual trimer assembly previously reported, but instead forms a canonical bell-shaped trimer typical of other TNF superfamily members. The structure of 4-1BB is also largely canonical as is the 4-1BB/4-1BBL complex. Mutational data support the 4-1BBL structure reported here as being biologically relevant, suggesting that the previously reported structure is not. Together, the data presented here offer insight into structure/function relationships in the 4-1BB/4-1BBL system and improve our structural understanding of the TNF/TNFR superfamily more broadly.
- Published
- 2018
19. Synergism interaction between genetic polymorphisms in drug metabolizing enzymes and NSAIDs on upper gastrointestinal haemorrhage: a multicenter case-control study
- Author
-
Narmeen Mallah, Maruxa Zapata-Cachafeiro, Carmelo Aguirre, Eguzkiñe Ibarra-García, Itziar Palacios-Zabalza, Fernando Macías-García, María Piñeiro-Lamas, Luisa Ibáñez, Xavier Vidal, Lourdes Vendrell, Luis Martin-Arias, María Sáinz-Gil, Verónica Velasco-González, Manuel Bacariza-Cortiñas, Angel Salgado, Ana Estany-Gestal, and Adolfo Figueiras
- Subjects
aspirin ,genetic variation ,interaction ,non-steroidal anti-inflammatory drugs ,upper gastrointestinal haemorrhage ,Medicine - Abstract
Background Interindividual genetic variations contribute to differences in patients’ response to drugs as well as to the development of certain disorders. Patients who use non-steroidal anti-inflammatory drugs (NSAIDs) may develop serious gastrointestinal disorders, mainly upper gastrointestinal haemorrhage (UGIH). Studies about the interaction between NSAIDs and genetic variations on the risk of UGIH are scarce. Therefore, we investigated the effect of 16 single nucleotide polymorphisms (SNPs) involved in drug metabolism on the risk of NSAIDs-induced UGIH. Materials and methods We conducted a multicenter case-control study of 326 cases and 748 controls. Participants were sub-grouped into four categories according to NSAID exposure and genetic profile. We estimated odds ratios (ORs) and their 95% confidence intervals (CI) using generalized linear mixed models for dependent binomial variables and then calculated the measures of interaction, synergism index (S), and relative excess risk due to interaction (RERI). We undertook stratified analyses by the type of NSAID (aspirin, non-aspirin). Results We observed an excess risk of UGIH due to an interaction between any NSAID, non-aspirin NSAIDs or aspirin and carrying certain SNPs. The greatest excess risk was observed for carriers of: rs2180314:C>G [any NSAID: S = 3.30 (95%CI: 1.24–8.80), RERI = 4.39 (95%CI: 0.70–8.07); non-aspirin NSAIDs: S = 3.42 (95%CI: 1.12–10.47), RERI = 3.97 (95%CI: 0.44–7.50)], and rs4809957:A>G [any NSAID: S = 2.11 (95%CI: 0.90–4.97), RERI = 3.46 (95%CI: −0.40–7.31)]. Aspirin use by carriers of rs6664:C>T is also associated with increased risk of UGIH [ORaspirin(+),wild-type: 2.22 (95%CI: 0.69–7.17) vs. ORaspirin(+),genetic-variation: 7.72 (95%CI: 2.75–21.68)], yet larger sample size is needed to confirm this observation. Conclusions The joint effect of the SNPs s2180314:C>G and rs4809957:A>G and NSAIDs are more than three times higher than the sum of their individual effects. Personalized prescriptions based on genotyping would permit a better weighing of risks and benefits from NSAID consumption.KEY MESSAGES Multicenter case-control study of the effect of genetic variations involved in drug metabolism on upper gastrointestinal haemorrhage (UGIH) induced by NSAIDs (aspirin and non-aspirin). There is a statistically significant additive synergism interaction between certain genetic polymorphisms and NSAIDs on UGIH: rs2180314:C>G and rs4809957:A>G. The joint effect of each of these single nucleotide polymorphisms and NSAIDs on UGIH is more than three times higher than the sum of their individual effects. Genetic profiling and personalized prescriptions would be useful in managing the risks and benefits associated with NSAIDs.
- Published
- 2022
- Full Text
- View/download PDF
20. Fibronectin type III domains engineered to bind CD40L: cloning, expression, purification, crystallization and preliminary X-ray diffraction analysis of two complexes
- Author
-
Luba Grinberg, Sandrina Phipps, Stacey Drabic, Thomas Thisted, Andrew D. Ferguson, Lin Wang, Vaheh Oganesyan, Benoy Chacko, and Manuel Baca
- Subjects
CD40 Ligand ,Molecular Sequence Data ,Biophysics ,Gene Expression ,Tenascin ,Sequence alignment ,Plasma protein binding ,Fibronectin type III domain ,Biology ,Crystallography, X-Ray ,Biochemistry ,Protein Structure, Secondary ,Peptide Library ,Structural Biology ,Escherichia coli ,Genetics ,Humans ,Amino Acid Sequence ,Binding site ,Peptide library ,Peptide sequence ,Binding Sites ,biochemical phenomena, metabolism, and nutrition ,Condensed Matter Physics ,Recombinant Proteins ,Fibronectins ,Fibronectin ,Crystallography ,Crystallization Communications ,biology.protein ,Peptides ,Sequence Alignment ,Protein Binding - Abstract
Tn3 proteins are a novel class of binding molecules based on the third fibronectin type III domain of human tenascin C. Target-specific Tn3 proteins are selected from combinatorial libraries in which three surface-exposed loops have been diversified. Here, the cocrystallization of two different Tn3 proteins in complex with CD40L, a therapeutic target for immunological disease, is reported. These crystal structures are the first to be reported of Tn3 proteins and will help to reveal how these engineered molecules achieve specific recognition of a cognate target.
- Published
- 2013
- Full Text
- View/download PDF
21. Multivalent Scaffold Proteins as Superagonists of TRAIL Receptor 2–Induced Apoptosis
- Author
-
Rosa A. Carrasco, Luba Grinberg, Lin Wang, Ching Ching Leow, Zhan Xiao, Kristen Lekstrom, Ivan Inigo, Herren Wu, Manuel Baca, Hui Feng, Jeffrey S. Swers, and David A. Tice
- Subjects
Agonist ,Scaffold protein ,Cancer Research ,Programmed cell death ,medicine.drug_class ,Recombinant Fusion Proteins ,Mice, Nude ,Apoptosis ,Biology ,Jurkat cells ,Jurkat Cells ,Mice ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Receptor ,Hep G2 Cells ,Ligand (biochemistry) ,Xenograft Model Antitumor Assays ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,Oncology ,Cell culture ,Immunology ,Cancer research ,Female - Abstract
Activation of TNF-related apoptosis-inducing ligand receptor 2 (TRAILR2) can induce apoptosis in a variety of human cancer cell lines and xenografts, while lacking toxicity in normal cells. The natural ligand and agonistic antibodies show antitumor activity in preclinical models of cancer, and this had led to significant excitement in the clinical potential of these agents. Unfortunately, this optimism has been tempered by trial data that, thus far, are not showing clear signs of efficacy in cancer patients. The reasons for discrepant preclinical and clinical observations are not understood, but one possibility is that the current TRAILR2 agonists lack sufficient potency to achieve a meaningful response in patients. Toward addressing that possibility, we have developed multivalent forms of a new binding scaffold (Tn3) that are superagonists of TRAILR2 and can induce apoptosis in tumor cell lines at subpicomolar concentrations. The monomer Tn3 unit was a fibronectin type III domain engineered for high-affinity TRAILR2 binding. Multivalent presentation of this basic unit induced cell death in TRAILR2-expressing cell lines. Optimization of binding affinity, molecular format, and valency contributed to cumulative enhancements of agonistic activity. An optimized multivalent agonist consisting of 8 tandem Tn3 repeats was highly potent in triggering cell death in TRAIL-sensitive cell lines and was 1 to 2 orders of magnitude more potent than TRAIL. Enhanced potency was also observed in vivo in a tumor xenograft setting. The TRAILR2 superagonists described here have the potential for superior clinical activity in settings insensitive to the current therapeutic agonists that target this pathway. Mol Cancer Ther; 12(7); 1235–44. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
22. A hingeless Fc fusion system for site-specific cleavage by IdeS
- Author
-
Manuel Baca, Keith W Rickert, Arnita Barnes, Shabazz Novarra, Luba Grinberg, and Susan Wilson
- Subjects
0301 basic medicine ,Proteases ,medicine.medical_treatment ,Recombinant Fusion Proteins ,Immunology ,Cleavage (embryo) ,Mass Spectrometry ,Substrate Specificity ,03 medical and health sciences ,0302 clinical medicine ,Bacterial Proteins ,Protein Domains ,Cleave ,Report ,medicine ,Immunology and Allergy ,Humans ,Receptor ,Hinge Exons ,chemistry.chemical_classification ,Protease ,Chemistry ,Fragment crystallizable region ,Fusion protein ,Immunoglobulin Fc Fragments ,030104 developmental biology ,Enzyme ,Biochemistry ,030220 oncology & carcinogenesis ,Immunoglobulin G ,Proteolysis ,Chromatography, Gel ,Chromatography, Liquid - Abstract
Fusion of proteins to the Fc region of IgG is widely used to express cellular receptors and other extracellular proteins, but cleavage of the fusion partner is sometimes required for downstream applications. Immunoglobulin G-degrading enzyme of Streptococcus pyogenes (IdeS) is a protease with exquisite specificity for human IgG, and it can also cleave Fc-fusion proteins at a single site in the N-terminal region of the CH2 domain. However, the site of IdeS cleavage results in the disulfide-linked hinge region partitioning with the released protein, complicating downstream usage of the cleaved product. To tailor the Fc fragment for release of partner proteins by IdeS treatment, we investigated the effect of deleting regions of IgG-derived sequence that are upstream of the cleavage site. Elimination of the IgG-derived hinge sequence along with several residues of the CH2 domain had negligible effects on expression and purity of the fusion protein, while retaining efficient processing by IdeS. An optimal Fc fragment comprising residues 235-447 of the human IgG1 heavy chain sufficed for efficient production of fusion proteins and minimized the amount of residual Ig-derived sequence on the cleavage product following IdeS treatment. Pairing of this truncated Fc fragment with IdeS cleavage enables highly specific cleavage of Fc-fusion proteins, thus eliminating the need to engineer extraneous cleavage sequences. This system should be helpful for producing Fc-fusion proteins requiring downstream cleavage, particularly those that are sensitive to internal miscleavage if treated with alternative proteases.
- Published
- 2016
23. Combining phage display with de novo protein sequencing for reverse engineering of monoclonal antibodies
- Author
-
Keith W Rickert, Manuel Baca, Michael A. Bowen, Robert M. Woods, Luba Grinberg, and Susan Wilson
- Subjects
0301 basic medicine ,Phage display ,medicine.drug_class ,Immunology ,Biology ,Proteomics ,Monoclonal antibody ,Protein Engineering ,03 medical and health sciences ,Mice ,Tumor Necrosis Factor Receptor Superfamily, Member 9 ,Protein sequencing ,Antigen ,Peptide Library ,Sequence Analysis, Protein ,medicine ,Immunology and Allergy ,Animals ,Peptide library ,Genetics ,Protein engineering ,Rats ,030104 developmental biology ,Single-Chain Antibodies ,Reports - Abstract
The enormous diversity created by gene recombination and somatic hypermutation makes de novo protein sequencing of monoclonal antibodies a uniquely challenging problem. Modern mass spectrometry-based sequencing will rarely, if ever, provide a single unambiguous sequence for the variable domains. A more likely outcome is computation of an ensemble of highly similar sequences that can satisfy the experimental data. This outcome can result in the need for empirical testing of many candidate sequences, sometimes iteratively, to identity one which can replicate the activity of the parental antibody. Here we describe an improved approach to antibody protein sequencing by using phage display technology to generate a combinatorial library of sequences that satisfy the mass spectrometry data, and selecting for functional candidates that bind antigen. This approach was used to reverse engineer 2 commercially-obtained monoclonal antibodies against murine CD137. Proteomic data enabled us to assign the majority of the variable domain sequences, with the exception of 3-5% of the sequence located within or adjacent to complementarity-determining regions. To efficiently resolve the sequence in these regions, small phage-displayed libraries were generated and subjected to antigen binding selection. Following enrichment of antigen-binding clones, 2 clones were selected for each antibody and recombinantly expressed as antigen-binding fragments (Fabs). In both cases, the reverse-engineered Fabs exhibited identical antigen binding affinity, within error, as Fabs produced from the commercial IgGs. This combination of proteomic and protein engineering techniques provides a useful approach to simplifying the technically challenging process of reverse engineering monoclonal antibodies from protein material.
- Published
- 2016
24. Endogenous IL-11 Signaling Is Essential in Th2- and IL-13–Induced Inflammation and Mucus Production
- Author
-
Felicity Meredith Dunlop, Chun Geun Lee, Hiroshi Matsuura, Jack A. Elias, Qingsheng Chen, Pierre Scotney, Dominik Hartl, Robert J. Homer, Louis Fabri, Chu-Yan Tang, Andrew D. Nash, Manuel Baca, and Ning Yuan Chen
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Ovalbumin ,Clinical Biochemistry ,Cell Count ,Inflammation ,Endogeny ,Mucin 5AC ,Immunoglobulin E ,Mice ,Th2 Cells ,Internal medicine ,medicine ,Animals ,Receptors, Interleukin-11 ,Receptor ,Molecular Biology ,Interleukin-13 ,biology ,Tumor Necrosis Factor-alpha ,Articles ,Cell Biology ,Allergens ,respiratory system ,Interleukin-11 ,Mucus ,Mice, Inbred C57BL ,Phenotype ,Endocrinology ,Gene Expression Regulation ,Interleukin 13 ,Immunology ,biology.protein ,Immunization ,Tumor necrosis factor alpha ,medicine.symptom ,Bronchoalveolar Lavage Fluid ,Signal Transduction - Abstract
IL-11 and IL-11 receptor (R)alpha are induced by Th2 cytokines. However, the role(s) of endogenous IL-11 in antigen-induced Th2 inflammation has not been fully defined. We hypothesized that IL-11, signaling via IL-11Ralpha, plays an important role in aeroallergen-induced Th2 inflammation and mucus metaplasia. To test this hypothesis, we compared the responses induced by the aeroallergen ovalbumin (OVA) in wild-type (WT) and IL-11Ralpha-null mutant mice. We also generated and defined the effects of an antagonistic IL-11 mutein on pulmonary Th2 responses. Increased levels of IgE, eosinophilic tissue and bronchoalveolar lavage (BAL) inflammation, IL-13 production, and increased mucus production and secretion were noted in OVA-sensitized and -challenged WT mice. These responses were at least partially IL-11 dependent because each was decreased in mice with null mutations of IL-11Ralpha. Importantly, the administration of the IL-11 mutein to OVA-sensitized mice before aerosol antigen challenge also caused a significant decrease in OVA-induced inflammation, mucus responses, and IL-13 production. Intraperitoneal administration of the mutein to lung-specific IL-13-overexpressing transgenic mice also reduced BAL inflammation and airway mucus elaboration. These studies demonstrate that endogenous IL-11R signaling plays an important role in antigen-induced sensitization, eosinophilic inflammation, and airway mucus production. They also demonstrate that Th2 and IL-13 responses can be regulated by interventions that manipulate IL-11 signaling in the murine lung.
- Published
- 2008
- Full Text
- View/download PDF
25. Correction for Zhang et al., The conserved SOCS box motif in suppressors of cytokine signaling binds to elongins B and C and may couple bound proteins to proteasomal degradation
- Author
-
Alison Farley, Nicos A. Nicola, Dale Cary, Donald Metcalf, Lisa M. Zugaro, Warren S. Alexander, Richard J. Simpson, Tracy A. Willson, Benjamin T. Kile, Rachael T. Richardson, Douglas J. Hilton, Robert L. Moritz, Stephen B. H. Kent, George Hausmann, Sandra E. Nicholson, Manuel Baca, and Jian-Guo Zhang
- Subjects
Proteasome Endopeptidase Complex ,medicine.medical_treatment ,Recombinant Fusion Proteins ,Zhàng ,Elongin ,Molecular Sequence Data ,Suppressor of Cytokine Signaling Proteins ,Computational biology ,Biology ,Bioinformatics ,Transfection ,Corrections ,law.invention ,Cell Line ,src Homology Domains ,Mice ,Suppressor of Cytokine Signaling 1 Protein ,law ,Multienzyme Complexes ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,Amino Acid Sequence ,Multidisciplinary ,Binding Sites ,Intracellular Signaling Peptides and Proteins ,Proteins ,Protein-Tyrosine Kinases ,Recombinant Proteins ,Repressor Proteins ,Cysteine Endopeptidases ,Cytokine ,Models, Chemical ,Suppressor of Cytokine Signaling 3 Protein ,Suppressor ,Cytokines ,Motif (music) ,Carrier Proteins ,Signal Transduction ,Transcription Factors - Abstract
The suppressors of cytokine signaling (SOCS) family of proteins act as intracellular inhibitors of several cytokine signal transduction pathways. Their expression is induced by cytokine activation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway and they act as a negative feedback loop by subsequently inhibiting the JAK/STAT pathway either by direct interaction with activated JAKs or with the receptors. These interactions are mediated at least in part by the SH2 domain of SOCS proteins but these proteins also contain a highly conserved C-terminal homology domain termed the SOCS box. Here we show that the SOCS box mediates interactions with elongins B and C, which in turn may couple SOCS proteins and their substrates to the proteasomal protein degradation pathway. Analogous to the family of F-box-containing proteins, it appears that the SOCS proteins may act as adaptor molecules that target activated cell signaling proteins to the protein degradation pathway.
- Published
- 2015
26. The Structure of SOCS3 Reveals the Basis of the Extended SH2 Domain Function and Identifies an Unstructured Insertion That Regulates Stability
- Author
-
Shenggen Yao, Manuel Baca, Tracy A. Willson, Lisa A. Mielke, David P. DeSouza, Sandra E. Nicholson, Raymond S. Norton, Edward J. McManus, Nicos A. Nicola, Naomi S. Sprigg, Douglas J. Hilton, and Jeffrey J. Babon
- Subjects
Phosphotyrosine binding ,Models, Molecular ,Amino Acid Motifs ,Molecular Sequence Data ,Suppressor of Cytokine Signaling Proteins ,Plasma protein binding ,Biology ,SH2 domain ,Spectrum Analysis, Raman ,Transfection ,Protein Structure, Secondary ,Cell Line ,src Homology Domains ,Protein structure ,EVH1 domain ,Genes, Reporter ,Humans ,Amino Acid Sequence ,Binding site ,Cloning, Molecular ,Luciferases ,Peptide sequence ,Nuclear Magnetic Resonance, Biomolecular ,Molecular Biology ,Binding Sites ,Sequence Homology, Amino Acid ,digestive, oral, and skin physiology ,Cell Biology ,Phosphoproteins ,Protein Structure, Tertiary ,Mutagenesis, Insertional ,Biochemistry ,Suppressor of Cytokine Signaling 3 Protein ,Biophysics ,Hydrophobic and Hydrophilic Interactions ,Binding domain ,Half-Life ,Protein Binding - Abstract
SOCS3 is essential for regulating the extent, duration, and specificity of cellular responses to cytokines such as G-CSF and IL-6. Here we describe the solution structure of SOCS3, the first structure determined for any SOCS protein, in complex with a phosphotyrosine-containing peptide from the IL-6 receptor signaling subunit gp130. The structure of the complex shows that seven peptide residues form a predominantly hydrophobic binding motif. Regions outside the SOCS3 SH2 domain are important for ligand binding, in particular, a single 15 residue alpha helix immediately N-terminal to the SH2 domain makes direct contacts with the phosphotyrosine binding loop and, in part, determines its geometry. The SH2 domain itself is remarkable in that it contains a 35 residue unstructured PEST motif insertion that is not required for STAT inhibition. The PEST motif increases SOCS3 turnover and affects its degradation pathway, implying that it has an important regulatory role inside the cell.
- Published
- 2006
- Full Text
- View/download PDF
27. Secondary structure assignment of mouse SOCS3 by NMR defines the domain boundaries and identifies an unstructured insertion in the SH2 domain
- Author
-
Sergio D. B. Scrofani, Raymond S. Norton, Christopher F. Harrison, David P. DeSouza, Jeffrey J. Babon, Manuel Baca, Shenggen Yao, Louis Fabri, and Edvards Liepinsh
- Subjects
Phosphotyrosine binding ,Activator (genetics) ,C-terminus ,Isothermal titration calorimetry ,Cell Biology ,Biology ,SH2 domain ,Biochemistry ,Suppressor of cytokine signalling ,Cell biology ,PEST sequence ,Molecular Biology ,Protein secondary structure - Abstract
SOCS3 is a negative regulator of cytokine signalling that inhibits Janus kinase-signal transduction and activator of transcription (JAK-STAT) mediated signal tranduction by binding to phosphorylated tyrosine residues on intracellular subunits of various cytokine receptors, as well as possibly the JAK proteins. SOCS3 consists of a short N-terminal sequence followed by a kinase inhibitory region, an extended SH2 domain and a C-terminal suppressor of cytokine signalling (SOCS) box. SOCS3 and the related protein, cytokine-inducible SH2-containing protein, are unique among the SOCS family of proteins in containing a region of mostly low complexity sequence, between the SH2 domain and the C-terminal SOCS box. Using NMR, we assigned and determined the secondary structure of a murine SOCS3 construct. The SH2 domain, unusually, consists of 140 residues, including an unstructured insertion of 35 residues. This insertion fits the criteria for a PEST sequence and is not required for phosphotyrosine binding, as shown by isothermal titration calorimetry. Instead, we propose that the PEST sequence has a functional role unrelated to phosphotyrosine binding, possibly mediating efficient proteolytic degradation of the protein. The latter half of the kinase inhibitory region and the entire extended SH2 subdomain form a single α-helix. The mapping of the true SH2 domain, and the location of its C terminus more than 50 residues further downstream than predicted by sequence homology, explains a number of previously unexpected results that have shown the importance of residues close to the SOCS box for phosphotyrosine binding.
- Published
- 2005
- Full Text
- View/download PDF
28. Synergetic effect between phases in MoVTe(Sb)NbO catalysts used for the oxidation of propane into acrylic acid
- Author
-
Manuel Baca, Jean-Marc M. Millet, Mimoun Aouine, and Jean-Luc Dubois
- Subjects
010405 organic chemistry ,Inorganic chemistry ,chemistry.chemical_element ,010402 general chemistry ,Heterogeneous catalysis ,01 natural sciences ,Catalysis ,0104 chemical sciences ,Propene ,chemistry.chemical_compound ,chemistry ,Antimony ,Phase (matter) ,Dehydrogenation ,Physical and Theoretical Chemistry ,Tellurium ,Bifunctional - Abstract
Results are reported concerning the synergetic effect observed in the oxidation of propane to acrylic acid over the orthorhombic M1 and hexagonal M2 phases present in the most active and selective MoVTe(Sb)NbO catalysts. The pure phases and phase mixtures containing either tellurium or antimony have been prepared and individually tested as catalysts. The results obtained confirm that the phase responsible for the catalytic properties of the efficient catalysts is phase M1, and that M2 is poorly active. Mechanical mixtures of the pure phases have also been prepared and tested. All of the catalysts have been characterized before and after the catalytic reaction by X-ray diffraction, X-ray photoelectron spectroscopy, and high-resolution electron microscopy with EDS analyses. Although the synergetic effect previously described (bifunctional catalysis with the oxidative dehydrogenation of propane molecules on the orthorhombic M1 phase and the subsequent oxidation of propene on the hexagonal M2 phase) was observed, another cause related to migration of tellurium from the M2 phase to the surface of the M1 phase was indicated. This migration should balance a loss of tellurium in the active phase occurring under the conditions of a catalytic test, but may also create new dehydrogenation sites for propene and/or anneal total oxidation sites. The hexagonal M2 phase would thus play a role of tellurium reservoir for the active M1 phase. This effect was not reversible and concerned only the tellurium. Antimony, which is less volatile, should not be lost by the active phase. Furthermore, it was shown not to diffuse at the surface of the phases.
- Published
- 2005
- Full Text
- View/download PDF
29. Fourier transform infrared spectroscopic study of surface acidity by pyridine adsorption on the M1 active phase of the MoVTe(Sb)NbO catalysts used in propane oxidation
- Author
-
Manuel Baca, Jean-Marc M. Millet, A. Pigamo, and Jean-Luc Dubois
- Subjects
010405 organic chemistry ,Process Chemistry and Technology ,Inorganic chemistry ,chemistry.chemical_element ,02 engineering and technology ,General Chemistry ,021001 nanoscience & nanotechnology ,01 natural sciences ,Catalysis ,0104 chemical sciences ,chemistry.chemical_compound ,Adsorption ,chemistry ,Antimony ,Propane ,Pyridine ,Lewis acids and bases ,0210 nano-technology ,Selectivity ,Acrylic acid - Abstract
The adsorption of pyridine on the surface of the M1 active phase of the MoVTe(Sb)O catalysts has been studied for the determination of the Bronsted and Lewis acid sites. The results obtained showed medium Lewis and Bronsted acidity for antimony containing phases and low Lewis and Bronsted acidity for tellurium ones. The incorporation of niobium in the M1 phases resulted in a decrease of both acidities. The samples characterized have been tested as catalysts in the oxidation of propane to acrylic acid. The results obtained allowed to propose correlations between the acidic properties and the catalytic selectivity. 2005 Elsevier B.V. All rights reserved.
- Published
- 2005
- Full Text
- View/download PDF
30. Bulk oxidation state of the different cationic elements in the MoVTe(Sb)NbO catalysts for oxidation or ammoxidation of propane
- Author
-
Jean-Marc M. Millet and Manuel Baca
- Subjects
010405 organic chemistry ,Process Chemistry and Technology ,Inorganic chemistry ,Oxide ,Vanadium ,chemistry.chemical_element ,010402 general chemistry ,01 natural sciences ,Catalysis ,0104 chemical sciences ,Crystallography ,chemistry.chemical_compound ,chemistry ,Antimony ,Molybdenum ,Oxidation state ,Partial oxidation ,Ammoxidation ,Stoichiometry - Abstract
Different spectroscopic techniques like XANES, ESR, XPS and Mossbauer spectroscopy have been used to determine the oxidation state of the various cations in the M1 and M2 phases of the MoVTe(Sb)NbO catalysts used for the oxidation or ammoxidation of propane. It was observed that the tellurium or antimony M 1o rM2 phases contained mainly Te IV , and Nb V. Molybdenum, vanadium and antimony were shown to be present as Mo VI and Mo V ,V V and V IV and Sb V and Sb III . The M1 phase which is responsible for the high efficiency of catalysts, corresponds to the total stoichiometry (AO)2� x(A2O)xM20O56 with A = Sb or Te, M = Mo, V and Nb and 0 � x � 1. It has a structure with hexagonal channels occupied by the A cations and oxides. This channel may contain an excess of oxygen which constitutes a reservoir for the catalysts and which likely plays a role in the reoxidation of the catalytic sites. The balance of the charges introduced by these oxides anions was achieved by the partial oxidation of Sb III to Sb V in M1(Sb) and by the oxidation of Mo V to Mo VI in M1(Te), Te remaining always Te IV as shown from Mossbauer spectroscopy data. The characterization of the solids after catalytic test showed very few changes in the solids structures and compositions. The study allowed concluding that the lone pair elements does not have only a role as constituents of the surface catalytic sites but also as bulk components to store oxygen in the hexagonal channels and contributes to its rapid diffusion to the surface. # 2004 Elsevier B.V. All rights reserved. reported as (Te2O)2M20O56 and (SbO)2M20O56 with M = Mo, V, Nb. The V/Mo and Nb/Mo ratios can vary, but are most frequently close to 0.3 and 0.1, respectively (4,5). The phase containing Te has an oxide over- stoichiometry while the one containing Sb, an oxide sub- stoichiometry, they may better be reported with the general composition (AO)2� x(A2O)xM20O56 with A = Te or Sb and 0 � x � 1. The first structure model for the M1 phases has been proposed in 2002 based on an isomorphism with the phase Cs0.7(Nb,W)5O14 (4). The structure of the M1 phase has been described as a corner sharing MO6 (M = Mo, V, Nb) octahedra network with tellurium or antimony cations and oxygen anions occupying sites in hexagonal channels formed by the octahedra. Besides hexagonal channels, pentagonal and heptagonal channels are present and oriented in the same direction. The heptagonal channels have been
- Published
- 2005
- Full Text
- View/download PDF
31. Affinity Maturation of Leukemia Inhibitory Factor and Conversion to Potent Antagonists of Signaling
- Author
-
Raymond S. Norton, Donald Metcalf, Sandra Mifsud, Shenggen Yao, Joanne E. McCoubrie, Nicos A. Nicola, Alessandro D. Uboldi, Erinna F. Lee, David P De Souza, Chunxiao C Wang, Manuel Baca, and W. Douglas Fairlie
- Subjects
Cell signaling ,Leukemia Inhibitory Factor Receptor alpha Subunit ,Receptors, OSM-LIF ,Leukemia inhibitory factor receptor ,medicine.disease_cause ,Leukemia Inhibitory Factor ,Biochemistry ,Affinity maturation ,Mice ,Antigens, CD ,Peptide Library ,Cytokine Receptor gp130 ,medicine ,Animals ,Humans ,Receptors, Cytokine ,Binding site ,Nuclear Magnetic Resonance, Biomolecular ,Molecular Biology ,Mutation ,Binding Sites ,Membrane Glycoproteins ,biology ,Interleukin-6 ,Oncostatin M ,Cell Biology ,Molecular biology ,biology.protein ,Leukemia inhibitory factor ,Signal Transduction - Abstract
Leukemia inhibitory factor (LIF)-induced cell signaling occurs following sequential binding to the LIF receptor alpha-chain (LIFR), then to the gp130 co-receptor used by all members of the interleukin-6 family of cytokines. By monovalently displaying human LIF on the surface of M13 phage and randomizing clusters of residues in regions predicted to be important for human LIFR binding, we have identified mutations, which lead to significant increases in affinity for binding to LIFR. Six libraries were constructed in which regions of 4-6 amino acids were randomized then panned against LIFR. Mutations identified in three distinct clusters, residues 53-57, 102-103, and 150-155, gave rise to proteins with significantly increased affinity for binding to both human and mouse LIFR. Combining the mutations for each of these regions further increased the affinity, such that the best mutants bound to human LIFR with1000-fold higher affinity than wild-type human LIF. NMR analysis indicated that the mutations did not alter the overall structure of the molecule relative to the native protein, although some local changes occurred in the vicinity of the substituted residues. Despite increases in LIFR binding affinity, these mutants did not show any increase in activity as agonists of LIF-induced proliferation of Ba/F3 cells expressing human LIFR and gp130 compared with wild-type LIF. Incorporation of two additional mutations (Q29A and G124R), which were found to abrogate cell signaling, led to the generation of highly potent antagonists of both human and murine LIF-induced bioactivity.
- Published
- 2004
- Full Text
- View/download PDF
32. Biological Evidence That SOCS-2 Can Act Either as an Enhancer or Suppressor of Growth Hormone Signaling
- Author
-
Phillip O. Morgan, Helene M. Martin, Louis Fabri, Nicos A. Nicola, Tracy A. Willson, Anne L. Thaus, Christopher J. Greenhalgh, Nils Billestrup, Jason Corbin, Rachel T Uren, Donald Metcalf, Douglas J. Hilton, Manuel Baca, Jian-Guo Zhang, and Warren S. Alexander
- Subjects
inorganic chemicals ,Genetically modified mouse ,medicine.medical_specialty ,Transgene ,medicine.medical_treatment ,Mice, Transgenic ,Suppressor of Cytokine Signaling Proteins ,Biology ,Biochemistry ,Suppressor of cytokine signalling ,Mice ,Internal medicine ,otorhinolaryngologic diseases ,medicine ,Animals ,SOCS3 ,Receptor ,Molecular Biology ,Proteins ,Receptors, Somatotropin ,Cell Biology ,Recombinant Proteins ,Cell biology ,DNA-Binding Proteins ,Repressor Proteins ,Endocrinology ,Cytokine ,Growth Hormone ,Trans-Activators ,Phosphorylation ,sense organs ,Signal transduction ,psychological phenomena and processes ,Protein Binding ,Signal Transduction - Abstract
Suppressor of cytokine signaling (SOCS)-2 is a member of a family of intracellular proteins implicated in the negative regulation of cytokine signaling. The generation of SOCS-2-deficient mice, which grow to one and a half times the size of their wild-type littermates, suggests that SOCS-2 may attenuate growth hormone (GH) signaling. In vitro studies indicate that, while SOCS-2 can inhibit GH action at low concentrations, at higher concentrations it may potentiate signaling. To determine whether a similar enhancement of signaling is observed in vivo or alternatively whether increased SOCS-2 levels repress growth in vivo, we generated and analyzed transgenic mice that overexpress SOCS-2 from a human ubiquitin C promoter. These mice are not growth-deficient and are, in fact, significantly larger than wild-type mice. The overexpressed SOCS-2 was found to bind to endogenous GH receptors in a number of mouse organs, while phosphopeptide binding studies with recombinant SOCS-2 defined phosphorylated tyrosine 595 on the GH receptor as the site of interaction. Together, the data implicate SOCS-2 as having dual effects on GH signaling in vivo.
- Published
- 2002
- Full Text
- View/download PDF
33. Stabilization of the third fibronectin type III domain of human tenascin-C through minimal mutation and rational design
- Author
-
Lea T. Grinberg, Jeffrey S. Swers, Benoy Chacko, Manuel Baca, and Ryan N. Gilbreth
- Subjects
Chemistry ,Protein Conformation ,Protein Stability ,Point mutation ,Molecular Sequence Data ,Rational design ,Bioengineering ,Sequence alignment ,Tenascin ,Computational biology ,Protein engineering ,Fibronectin type III domain ,Protein Engineering ,Biochemistry ,Fibronectins ,Protein Structure, Tertiary ,Protein structure ,Mutation (genetic algorithm) ,Humans ,Point Mutation ,Amino Acid Sequence ,Binding site ,Molecular Biology ,Sequence Alignment ,Biotechnology - Abstract
Non-antibody scaffolds are increasingly used to generate novel binding proteins for both research and therapeutic applications. Our group has developed the tenth fibronectin type III domain of human tenascin-C (TNfn3) as one such scaffold. As a scaffold, TNfn3 must tolerate extensive mutation to introduce novel binding sites. However, TNfn3's marginal stability (T(m) ∼ 59°C, ΔG(unfolding) = 5.7 kcal/mol) stands as a potential obstacle to this process. To address this issue, we sought to engineer highly stable TNfn3 variants. We used two parallel strategies. Using insights gained from structural analysis of other FN3 family members, we (1) rationally designed stabilizing point mutations or (2) introduced novel stabilizing disulfide bonds. Both strategies yielded highly stable TNfn3 variants with T(m) values as high as 83°C and ΔG(unfolding) values as high as 9.4 kcal/mol. Notably, only three or four mutations were required to achieve this level of stability with either approach. These results validate our rational design strategies and illustrate that substantial stability increases can be achieved with minimal mutation. One TNfn3 variant reported here has now been successfully used as a scaffold to develop two promising therapeutic molecules. We anticipate that other variants described will exhibit similar utility.
- Published
- 2014
34. Cloning and characterization of the genes encoding the ankyrin repeat and SOCS box-containing proteins Asb-1, Asb-2, Asb-3 and Asb-4
- Author
-
Elizabeth M. Viney, Michael Cancilla, Benjamin T. Kile, Thomas C. Brodnicki, Tracy A. Willson, Douglas J. Hilton, Manuel Baca, Warren S. Alexander, Nicos A. Nicola, Ben A. Croker, and Amy S. Herlihy
- Subjects
Male ,inorganic chemicals ,DNA, Complementary ,Protein family ,Molecular Sequence Data ,Gene Expression ,Mice, Inbred Strains ,Biology ,SH2 domain ,behavioral disciplines and activities ,Suppressor of cytokine signalling ,Homology (biology) ,Mice ,otorhinolaryngologic diseases ,Genetics ,Animals ,Humans ,Tissue Distribution ,Amino Acid Sequence ,RNA, Messenger ,Cloning, Molecular ,Receptor ,Gene ,Base Sequence ,Sequence Homology, Amino Acid ,Kinase ,Chromosome Mapping ,DNA ,Exons ,Sequence Analysis, DNA ,General Medicine ,Blotting, Northern ,Molecular biology ,Introns ,Ankyrin Repeat ,Cell biology ,Mice, Inbred C57BL ,Genes ,Ankyrin repeat ,sense organs ,Carrier Proteins ,Sequence Alignment ,psychological phenomena and processes - Abstract
Members of the suppressor of cytokine signalling (SOCS) family of proteins have been shown to inhibit cytokine signalling via direct interactions with JAK kinases or activated cytokine receptors. In addition to their novel amino-terminal regions and SH2 domains that mediate these interactions, the SOCS proteins also contain carboxy-terminal regions of homology called the SOCS box. The SOCS box serves to couple SOCS proteins and their binding partners with the elongin B and C complex, possibly targeting them for degradation. Several other families of proteins also contain SOCS boxes but differ from the SOCS proteins in the type of domain or motif they contain upstream of the SOCS box. We report here the cloning, characterization, mapping and expression analysis of four members of the ankyrin repeat and SOCS box-containing (Asb) protein family.
- Published
- 2000
- Full Text
- View/download PDF
35. The conserved SOCS box motif in suppressors of cytokine signaling binds to elongins B and C and may couple bound proteins to proteasomal degradation
- Author
-
Richard J. Simpson, Benjamin Kile, Warren S. Alexander, George Hausmann, Tracy A. Willson, Manuel Baca, Donald Metcalf, Jian-Guo Zhang, Stephen B. H. Kent, Robert L. Moritz, Alison Farley, Sandra E. Nicholson, Douglas J. Hilton, Dale Cary, Nicos A. Nicola, Rachael T. Richardson, and Lisa M. Zugaro
- Subjects
Cell signaling ,Multidisciplinary ,otorhinolaryngologic diseases ,STAT protein ,JAK-STAT signaling pathway ,Signal transduction ,Biology ,Protein degradation ,Janus kinase ,SH2 domain ,Suppressor of cytokine signalling ,Cell biology - Abstract
The suppressors of cytokine signaling (SOCS) family of proteins act as intracellular inhibitors of several cytokine signal transduction pathways. Their expression is induced by cytokine activation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway and they act as a negative feedback loop by subsequently inhibiting the JAK/STAT pathway either by direct interaction with activated JAKs or with the receptors. These interactions are mediated at least in part by the SH2 domain of SOCS proteins but these proteins also contain a highly conserved C-terminal homology domain termed the SOCS box. Here we show that the SOCS box mediates interactions with elongins B and C, which in turn may couple SOCS proteins and their substrates to the proteasomal protein degradation pathway. Analogous to the family of F-box-containing proteins, it appears that the SOCS proteins may act as adaptor molecules that target activated cell signaling proteins to the protein degradation pathway.
- Published
- 1999
- Full Text
- View/download PDF
36. A Therapeutic Uricase with Reduced Immunogenicity Risk and Improved Development Properties
- Author
-
Flaviu Gruia, Jeffrey N. Miner, Frank Bartnik, David M. Wilson, Luba Grinberg, Ryan Bean, Jane K. Osbourn, Herren Wu, Hui Feng, Lutz Jermutus, James C. Geoghegan, Benoy Chacko, Vidyashankara Iyer, Susan Wilson, David A. Owen, Manuel Baca, Simone M Nicholson, Varnika Roy, Mark Berge, Anna Zacco, Steven Coats, Dongmei Zhou, Ellen O'Connor, Chris Ward, Chi Y. Wu, Michaela Wendeler, Andrew C. Nyborg, and Clynn Wilker
- Subjects
0301 basic medicine ,Gout ,Urate Oxidase ,Physiology ,Swine ,T-Lymphocytes ,lcsh:Medicine ,Pharmacology ,Biochemistry ,Polyethylene Glycols ,Substrate Specificity ,White Blood Cells ,Database and Informatics Methods ,chemistry.chemical_compound ,Subcutaneous injection ,Animal Cells ,Immune Physiology ,Medicine and Health Sciences ,Post-Translational Modification ,lcsh:Science ,chemistry.chemical_classification ,Immune System Proteins ,Multidisciplinary ,Calorimetry, Differential Scanning ,T Cells ,Immunogenicity ,Urate oxidase ,Hematology ,Hydrogen-Ion Concentration ,Recombinant Proteins ,Body Fluids ,Actinobacteria ,Blood ,Cellular Types ,Anatomy ,Sequence Analysis ,Research Article ,Half-Life ,Bioinformatics ,Immune Cells ,Inflammatory Diseases ,Immunology ,Research and Analysis Methods ,03 medical and health sciences ,Dogs ,Rheumatology ,Sequence Motif Analysis ,In vivo ,Escherichia coli ,medicine ,Animals ,Humans ,Arthrobacter ,Antigens ,Blood Cells ,Bacteria ,030102 biochemistry & molecular biology ,lcsh:R ,Organisms ,Biology and Life Sciences ,Proteins ,Cell Biology ,Pegylation ,medicine.disease ,Rats ,Kinetics ,030104 developmental biology ,Enzyme ,chemistry ,PEGylation ,Uric acid ,lcsh:Q ,Sequence Alignment ,Papio - Abstract
Humans and higher primates are unique in that they lack uricase, the enzyme capable of oxidizing uric acid. As a consequence of this enzyme deficiency, humans have high serum uric acid levels. In some people, uric acid levels rise above the solubility limit resulting in crystallization in joints, acute inflammation in response to those crystals causes severe pain; a condition known as gout. Treatment for severe gout includes injection of non-human uricase to reduce serum uric acid levels. Krystexxa® is a hyper-PEGylated pig-baboon chimeric uricase indicated for chronic refractory gout that induces an immunogenic response in 91% of treated patients, including infusion reactions (26%) and anaphylaxis (6.5%). These properties limit its use and effectiveness. An innovative approach has been used to develop a therapeutic uricase with improved properties such as: soluble expression, neutral pH solubility, high E. coli expression level, thermal stability, and excellent activity. More than 200 diverse uricase sequences were aligned to guide protein engineering and reduce putative sequence liabilities. A single uricase lead candidate was identified, which showed low potential for immunogenicity in >200 human donor samples selected to represent diverse HLA haplotypes. Cysteines were engineered into the lead sequence for site specific PEGylation and studies demonstrated >95% PEGylation efficiency. PEGylated uricase retains enzymatic activity in vitro at neutral pH, in human serum and in vivo (rats and canines) and has an extended half-life. In canines, an 85% reduction in serum uric acid levels was observed with a single subcutaneous injection. This PEGylated, non-immunogenic uricase has the potential to provide meaningful benefits to patients with gout.
- Published
- 2016
- Full Text
- View/download PDF
37. Structural engineering of the HIV-1 protease molecule with aβ-turn mimic of fixed geometry
- Author
-
Manuel Baca, Paul F. Alewood, and Stephen B. H. Kent
- Subjects
Models, Molecular ,Hot Temperature ,Stereochemistry ,Protein subunit ,medicine.medical_treatment ,Molecular Sequence Data ,Protein design ,Geometry ,Protein Engineering ,Peptides, Cyclic ,Biochemistry ,Protein Structure, Secondary ,Substrate Specificity ,Turn (biochemistry) ,HIV Protease ,HIV-1 protease ,Enzyme Stability ,medicine ,Amino Acid Sequence ,Cysteine ,Molecular Biology ,Protein secondary structure ,chemistry.chemical_classification ,Protease ,biology ,Dipeptides ,Amino acid ,Enzyme ,chemistry ,biology.protein ,Research Article - Abstract
An important goal in the de novo design of enzymes is the control of molecular geometry. To this end, an analog of the protease from human immunodeficiency virus 1 (HIV-1 protease) was prepared by total chemical synthesis, containing a constrained, nonpeptidic type II' beta-turn mimic of predetermined three-dimensional structure. The mimic beta-turn replaced residues Gly16,17 in each subunit of the homodimeric molecule. These residues constitute the central amino acids of two symmetry-related type I' beta-turns in the native, unliganded enzyme. The beta-turn mimic-containing enzyme analog was fully active, possessed the same substrate specificity as the Gly16,17-containing enzyme, and showed enhanced resistance to thermal inactivation. These results indicate that the precise geometry of the beta-turn at residues 15-18 in each subunit is not critical for activity, and that replacement of the native sequence with a rigid beta-turn mimic can lead to enhanced protein stability. Finally, the successful incorporation of a fixed element of secondary structure illustrates the potential of a "molecular kit set" approach to protein design and synthesis.
- Published
- 1993
- Full Text
- View/download PDF
38. Blocking LIF action in the uterus by using a PEGylated antagonist prevents implantation: a nonhormonal contraceptive strategy
- Author
-
Nicos A. Nicola, Jian-Guo Zhang, Lorraine Robb, Donald Metcalf, Christine A White, Lois A. Salamonsen, Manuel Baca, Evdokia Dimitriadis, and W. Douglas Fairlie
- Subjects
STAT3 Transcription Factor ,medicine.medical_specialty ,Uterus ,Biology ,Endometrium ,Leukemia Inhibitory Factor ,Polyethylene Glycols ,Mice ,Pharmacokinetics ,Pregnancy ,Internal medicine ,medicine ,Contraceptive Agents, Female ,Animals ,Blastocyst ,Embryo Implantation ,Phosphorylation ,STAT3 ,reproductive and urinary physiology ,Multidisciplinary ,Antagonist ,Biological Sciences ,medicine.disease ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Endocrinology ,Contraception ,embryonic structures ,biology.protein ,Female ,Leukemia inhibitory factor - Abstract
Blastocyst implantation is a critical stage in the establishment of pregnancy. Leukemia inhibitory factor (LIF) is essential for mouse blastocyst implantation and also plays a role in human pregnancy. We examined the effect of a potent LIF antagonist (LA) on mouse implantation. In mice, LIF expression peaks on day 3.5 of pregnancy (D3.5) (D0.5 = day of mating plug detection) in the uterine glandular epithelium. LA (7 mg/kg per day) administered from D2.5 to D4.5 via four hourly i.p. injections plus continuous administration via miniosmotic pump resulted in complete implantation failure. To improve its pharmacokinetic properties, we conjugated LA to polyethylene glycol (PEG), achieving a significant increase in serum levels. PEGylated LA (PEGLA) (37.5 mg/kg per day) administered via three i.p. injections between D2.5 and D3.5 also resulted in complete implantation failure. PEGLA immunolocalized to the uterine luminal epithelium at the time of blastocyst implantation. Both LA and PEGLA reduced phosphorylation of the downstream signaling molecule STAT3 in luminal epithelial cells on D3.5. The effects of PEGLA were found to be endometrial, with no embryo-lethal effects observed. These data demonstrate that administration of a PEGylated LIF antagonist is an effective method of targeting LIF signaling in the endometrium and a promising novel approach in the development of nonhormonal contraceptives for women.
- Published
- 2007
39. Secondary structure assignment of mouse SOCS3 by NMR defines the domain boundaries and identifies an unstructured insertion in the SH2 domain
- Author
-
Jeffrey J, Babon, Shenggen, Yao, David P, DeSouza, Christopher F, Harrison, Louis J, Fabri, Edvards, Liepinsh, Sergio D, Scrofani, Manuel, Baca, and Raymond S, Norton
- Subjects
Molecular Sequence Data ,Suppressor of Cytokine Signaling Proteins ,Protein Structure, Secondary ,src Homology Domains ,Mice ,Suppressor of Cytokine Signaling 3 Protein ,Animals ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Phosphotyrosine ,Nuclear Magnetic Resonance, Biomolecular ,Sequence Alignment ,Signal Transduction - Abstract
SOCS3 is a negative regulator of cytokine signalling that inhibits Janus kinase-signal transduction and activator of transcription (JAK-STAT) mediated signal tranduction by binding to phosphorylated tyrosine residues on intracellular subunits of various cytokine receptors, as well as possibly the JAK proteins. SOCS3 consists of a short N-terminal sequence followed by a kinase inhibitory region, an extended SH2 domain and a C-terminal suppressor of cytokine signalling (SOCS) box. SOCS3 and the related protein, cytokine-inducible SH2-containing protein, are unique among the SOCS family of proteins in containing a region of mostly low complexity sequence, between the SH2 domain and the C-terminal SOCS box. Using NMR, we assigned and determined the secondary structure of a murine SOCS3 construct. The SH2 domain, unusually, consists of 140 residues, including an unstructured insertion of 35 residues. This insertion fits the criteria for a PEST sequence and is not required for phosphotyrosine binding, as shown by isothermal titration calorimetry. Instead, we propose that the PEST sequence has a functional role unrelated to phosphotyrosine binding, possibly mediating efficient proteolytic degradation of the protein. The latter half of the kinase inhibitory region and the entire extended SH2 subdomain form a single alpha-helix. The mapping of the true SH2 domain, and the location of its C terminus more than 50 residues further downstream than predicted by sequence homology, explains a number of previously unexpected results that have shown the importance of residues close to the SOCS box for phosphotyrosine binding.
- Published
- 2005
40. Relaxin and prostaglandin E(2) regulate interleukin 11 during human endometrial stromal cell decidualization
- Author
-
Joanne E. McCoubrie, Lois A. Salamonsen, Evdokia Dimitriadis, C. Stoikos, W. D. Fairlie, and Manuel Baca
- Subjects
medicine.medical_specialty ,Stromal cell ,Swine ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Clinical Biochemistry ,Indomethacin ,Cell Culture Techniques ,Biology ,Biochemistry ,Dinoprostone ,Endometrium ,Endocrinology ,Internal medicine ,medicine ,Cyclic AMP ,Decidua ,Animals ,Humans ,Secretion ,Prostaglandin E2 ,Enzyme Inhibitors ,Endometrial Stromal Cell ,Relaxin ,Reverse Transcriptase Polymerase Chain Reaction ,Biochemistry (medical) ,Decidualization ,Thionucleotides ,Interleukin-11 ,Interleukin 11 ,Female ,Stromal Cells ,Prostaglandin E ,medicine.drug - Abstract
Decidualization of endometrial stromal cells and IL-11 signaling are essential for embryo implantation in the mouse. We investigated the effects of relaxin (RLX) and prostaglandin E(2) (PGE(2)) on IL-11 secretion by human endometrial stromal cells (HESC) and during cAMP or medroxyprogesterone acetate (P)-induced decidualization. cAMP-decidualized HESC secreted high levels of IL-11. RLX, cAMP, or PGE(2) increased IL-11 mRNA and IL-11 secretion, with maximal response to RLX and cAMP. Addition of the cAMP/protein kinase A inhibitor Rp-adenosine-3,5-cyclic-monophosphorothioate to either RLX- or PGE(2)-treated cells decreased IL-11 secretion. Indomethacin treatment decreased IL-11 secretion, which was largely restored by cotreatment with PGE(2) or RLX. Cotreatment of HESC with RLX, PGE(2), or cAMP and estrogen plus P down-regulated IL-11 mRNA and IL-11 secretion at 24 h, before secretion of prolactin (decidualization marker). Addition of W147AIL-11 (IL-11 signaling inhibitor) reduced prolactin secretion stimulated by RLX or PGE(2) and estrogen plus P. This is the first demonstration that cAMP-decidualized HESC secrete IL-11 and that IL-11 mRNA and IL-11 secretion are regulated by RLX and PGE(2), partly via a cAMP/protein kinase A-dependent pathway. Blocking IL-11 signaling reduced RLX+P- or PGE(2)+P-induced decidualization, suggesting that RLX and PGE(2) act via IL-11. This is important in understanding implantation and regulation of fertility.
- Published
- 2005
41. Negative regulation of gp130 signalling mediated through tyrosine-757 is not dependent on the recruitment of SHP2
- Author
-
David P De Souza, Manuel Baca, Nicos A. Nicola, and W. Douglas Fairlie
- Subjects
Gene Expression ,Protein Tyrosine Phosphatase, Non-Receptor Type 11 ,Suppressor of Cytokine Signaling Proteins ,SH2 domain ,Ligands ,Biochemistry ,Mice ,Cytokine Receptor gp130 ,Receptors, Erythropoietin ,Protein Phosphatase 2 ,Phosphorylation ,Luciferases ,Promoter Regions, Genetic ,SOCS2 ,Cells, Cultured ,Membrane Glycoproteins ,digestive, oral, and skin physiology ,Intracellular Signaling Peptides and Proteins ,Cell biology ,Interleukin-21 receptor ,Proto-oncogene tyrosine-protein kinase Src ,Plasmids ,Protein Binding ,Signal Transduction ,Research Article ,SH2 Domain-Containing Protein Tyrosine Phosphatases ,Recombinant Fusion Proteins ,Blotting, Western ,Biology ,Transfection ,Suppressor of cytokine signalling ,src Homology Domains ,Antigens, CD ,Animals ,Humans ,Molecular Biology ,Common gamma chain ,Interleukin-6 ,Proteins ,Cell Biology ,Fibroblasts ,Interleukin-13 receptor ,Glycoprotein 130 ,Precipitin Tests ,Repressor Proteins ,Suppressor of Cytokine Signaling 3 Protein ,Mutation ,Mutagenesis, Site-Directed ,Tyrosine ,Protein Tyrosine Phosphatases ,Transcription Factors - Abstract
Cytokines of the interleukin-6 family utilize the shared cytokine receptor gp130 in the formation of active signalling complexes. Tyrosine-757 (Y757) on this receptor is critical for negative regulation of gp130-mediated signalling. Two signalling regulators, suppressor of cytokine signalling 3 (SOCS3) and Src homology 2 domain-containing tyrosine phosphatase-2 (SHP2), are recruited to Y757 following receptor activation; however, the relative contribution made by each of these in down-regulating gp130 signalling is not known. In the present study, we show the design of a mutant gp130 receptor that can recruit SHP2, but not SOCS3. This receptor maintains the critical Y757 residue, but contains mutations in other surrounding residues which are also important for interactions with the Src homology 2 domains of SOCS3 and SHP2. Cells transfected with a chimaeric receptor containing the SHP2-selective gp130 intracellular domain showed an enhanced response to cytokine stimulation, which was similar to that shown by a chimaeric gp130 receptor mutant carrying a Y757F point mutation that failed to recruit either SOCS3 or SHP2. These results demonstrate that the recruitment of SHP2 alone is not sufficient for Y757-dependent negative regulation of gp130 signalling and that this activity must therefore be dependent on SOCS3.
- Published
- 2003
42. SOCS-6 binds to insulin receptor substrate 4, and mice lacking the SOCS-6 gene exhibit mild growth retardation
- Author
-
Robyn Starr, Richard J. Simpson, Donald Metcalf, Jian-Guo Zhang, Lisa M. Connolly, Kathy Hanzinikolas, Nicos A. Nicola, Manuel Baca, Douglas J. Hilton, David P De Souza, Danielle L. Krebs, Rachel T Uren, Jo L. Eyles, Steven Rakar, Warren S. Alexander, and Sandra E. Nicholson
- Subjects
inorganic chemicals ,Blood Glucose ,Male ,Protein subunit ,Hematopoietic System ,Elongin ,Growth ,Biology ,SH2 domain ,behavioral disciplines and activities ,src Homology Domains ,chemistry.chemical_compound ,Mice ,Phosphatidylinositol 3-Kinases ,Ubiquitin ,Insulin Receptor Substrate 4 ,otorhinolaryngologic diseases ,Mammalian Genetic Models with Minimal or Complex Phenotypes ,Glucose homeostasis ,Animals ,Insulin ,Phosphatidylinositol ,Molecular Biology ,Binding Sites ,Intracellular Signaling Peptides and Proteins ,Proteins ,Cell Biology ,Phosphoproteins ,Molecular biology ,Mice, Mutant Strains ,Ubiquitin ligase ,Mice, Inbred C57BL ,chemistry ,biology.protein ,Insulin Receptor Substrate Proteins ,Body Constitution ,Female ,sense organs ,Signal transduction ,psychological phenomena and processes ,Transcription Factors - Abstract
SOCS-6 is a member of the suppressor of cytokine signaling (SOCS) family of proteins (SOCS-1 to SOCS-7 and CIS) which each contain a central SH2 domain and a carboxyl-terminal SOCS box. SOCS-1, SOCS-2, SOCS-3, and CIS act to negatively regulate cytokine-induced signaling pathways; however, the actions of SOCS-4, SOCS-5, SOCS-6, and SOCS-7 remain less clear. Here we have used both biochemical and genetic approaches to examine the action of SOCS-6. We found that SOCS-6 and SOCS-7 are expressed ubiquitously in murine tissues. Like other SOCS family members, SOCS-6 binds to elongins B and C through its SOCS box, suggesting that it might act as an E3 ubiquitin ligase that targets proteins bound to its SH2 domain for ubiquitination and proteasomal degradation. We investigated the binding specificity of the SOCS-6 and SOCS-7 SH2 domains and found that they preferentially bound to phosphopeptides containing a valine in the phosphotyrosine (pY) +1 position and a hydrophobic residue in the pY +2 and pY +3 positions. In addition, these SH2 domains interacted with a protein complex consisting of insulin receptor substrate 4 (IRS-4), IRS-2, and the p85 regulatory subunit of phosphatidylinositol 3-kinase. To investigate the physiological role of SOCS-6, we generated mice lacking the SOCS-6 gene. SOCS-6(-/-) mice were born in a normal Mendelian ratio, were fertile, developed normally, and did not exhibit defects in hematopoiesis or glucose homeostasis. However, both male and female SOCS-6(-/-) mice weighed approximately 10% less than wild-type littermates.
- Published
- 2002
43. Direct inhibition of caspase 3 is dispensable for the anti-apoptotic activity of XIAP
- Author
-
Christine J. Hawkins, Joanne Chew, Paul G Ekert, Manuel Baca, John Silke, Anne M. Verhagen, Miha Pakusch, David L. Vaux, and Catherine L. Day
- Subjects
Ultraviolet Rays ,Molecular Sequence Data ,Gene Expression ,Caspase 3 ,Apoptosis ,X-Linked Inhibitor of Apoptosis Protein ,Inhibitor of apoptosis ,General Biochemistry, Genetics and Molecular Biology ,Article ,Inhibitor of Apoptosis Proteins ,Mitochondrial Proteins ,Viral Proteins ,Schizosaccharomyces ,Animals ,Amino Acid Sequence ,Molecular Biology ,Caspase ,Caspase-9 ,General Immunology and Microbiology ,biology ,General Neuroscience ,Proteins ,biology.organism_classification ,Molecular biology ,Caspase Inhibitors ,Caspase 9 ,XIAP ,Enzyme Activation ,Mutagenesis ,Caspases ,biology.protein ,Baculoviral IAP repeat-containing protein 3 ,Carrier Proteins - Abstract
XIAP is a mammalian inhibitor of apoptosis protein (IAP). To determine residues within the second baculoviral IAP repeat (BIR2) required for inhibition of caspase 3, we screened a library of BIR2 mutants for loss of the ability to inhibit caspase 3 toxicity in the yeast Schizosaccharomyces pombe. Four of the mutations, not predicted to affect the structure of the BIR fold, clustered together on the N-terminal region that flanks BIR2, suggesting that this is a site of interaction with caspase 3. Introduction of these mutations into full-length XIAP reduced caspase 3 inhibitory activity up to 500-fold, but did not affect its ability to inhibit caspase 9 or interact with the IAP antagonist DIABLO. Furthermore, these mutants retained full ability to inhibit apoptosis in transfected cells, demonstrating that although XIAP is able to inhibit caspase 3, this activity is dispensable for inhibition of apoptosis by XIAP in vivo.
- Published
- 2001
44. Chopper, a new death domain of the p75 neurotrophin receptor that mediates rapid neuronal cell death
- Author
-
Elizabeth J. Coulson, Perry F. Bartlett, Kate Reid, Kylie Shipham, Sarah M. Hulett, Manuel Baca, and Trevor J. Kilpatrick
- Subjects
Programmed cell death ,Receptors, Nerve Growth Factor ,Biochemistry ,Receptors, Tumor Necrosis Factor ,Chopper ,Antigens, CD ,Low-affinity nerve growth factor receptor ,Receptors, Tumor Necrosis Factor, Type II ,Molecular Biology ,Caspase ,Death domain ,Neurons ,biology ,Cell Death ,Calpain ,Cell Polarity ,Membrane Proteins ,Receptor Protein-Tyrosine Kinases ,Cell Biology ,Peptide Fragments ,Cell biology ,Protein Structure, Tertiary ,Neuroprotective Agents ,Ectodomain ,Cytoplasm ,Trk receptor ,Caspases ,biology.protein ,sense organs ,Signal Transduction - Abstract
The cytoplasmic juxtamembrane region of the p75 neurotrophin receptor (p75NTR) has been found to be necessary and sufficient to initiate neural cell death. The region was named “Chopper” to distinguish it from CD95-like death domains. A 29-amino acid peptide corresponding to the Chopper region induced caspase- and calpain-mediated death in a variety of neural and non-neural cell types and was not inhibited by signaling through Trk (unlike killing by full-length p75NTR). Chopper triggered cell death only when bound to the plasma membrane by a lipid anchor, whereas non-anchored Chopper acted in a dominant-negative manner, blocking p75NTR-mediated death both in vitroand in vivo. Removal of the ectodomain of p75NTR increased the potency of Chopper activity, suggesting that it regulates the association of Chopper with downstream signaling proteins.
- Published
- 2000
45. Suppressor of cytokine signaling-3 preferentially binds to the SHP-2-binding site on the shared cytokine receptor subunit gp130
- Author
-
Maria Asimakis, Jason Corbin, Sandra E. Nicholson, Manuel Baca, Tracy A. Willson, Anabel Silva, Jian-Guo Zhang, Louis Fabri, Alison Farley, Andrew D. Nash, David P De Souza, Douglas J. Hilton, Donald Metcalf, and Nicos A. Nicola
- Subjects
SH2 Domain-Containing Protein Tyrosine Phosphatases ,Molecular Sequence Data ,Protein Tyrosine Phosphatase, Non-Receptor Type 11 ,Suppressor of Cytokine Signaling Proteins ,Biology ,Suppressor of cytokine signalling ,Antigens, CD ,otorhinolaryngologic diseases ,Cytokine Receptor gp130 ,Amino Acid Sequence ,SOCS2 ,Common gamma chain ,Multidisciplinary ,Binding Sites ,Membrane Glycoproteins ,Janus kinase 1 ,Suppressor of cytokine signaling 1 ,Protein Tyrosine Phosphatase, Non-Receptor Type 6 ,Intracellular Signaling Peptides and Proteins ,Proteins ,Janus Kinase 1 ,Protein-Tyrosine Kinases ,Biological Sciences ,Interleukin-13 receptor ,Glycoprotein 130 ,Molecular biology ,Cell biology ,Repressor Proteins ,Suppressor of Cytokine Signaling 3 Protein ,biological phenomena, cell phenomena, and immunity ,Protein Tyrosine Phosphatases ,Cytokine receptor ,Signal Transduction ,Transcription Factors - Abstract
Suppressor of cytokine signaling-3 (SOCS-3) is one member of a family of intracellular inhibitors of signaling pathways initiated by cytokines that use, among others, the common receptor subunit gp130. The SH2 domain of SOCS-3 has been shown to be essential for this inhibitory activity, and we have used a quantitative binding analysis of SOCS-3 to synthetic phosphopeptides to map the potential sites of interaction of SOCS-3 with different components of the gp130 signaling pathway. The only high-affinity ligand found corresponded to the region of gp130 centered around phosphotyrosine-757 (pY757), previously shown to be a docking site for the tyrosine phosphatase SHP-2. By contrast, phosphopeptides corresponding to other regions within gp130, Janus kinase, or signal transducer and activator of transcription proteins bound to SOCS-3 with weak or undetectable affinity. The significance of pY757 in gp130 as a biologically relevant SOCS-3 docking site was investigated by using transfected 293T fibroblasts. Although SOCS-3 inhibited signaling in cells transfected with a chimeric receptor containing the wild-type gp130 intracellular domain, inhibition was considerably impaired for a receptor carrying a Y→F point mutation at residue 757. Taken together, these data suggest that the mechanism by which SOCS-3 inhibits the gp130 signaling pathway depends on recruitment to the phosphorylated gp130 receptor, and that some of the negative regulatory roles previously attributed to the phosphatase SHP-2 might in fact be caused by the action of SOCS-3.
- Published
- 2000
46. Mutational analyses of the SOCS proteins suggest a dual domain requirement but distinct mechanisms for inhibition of LIF and IL-6 signal transduction
- Author
-
Tracy A. Willson, Jian-Guo Zhang, Donald Metcalf, Sandra E. Nicholson, Robyn Starr, Douglas J. Hilton, Manuel Baca, Nicos A. Nicola, Alison Farley, and Warren S. Alexander
- Subjects
inorganic chemicals ,Cellular differentiation ,Suppressor of Cytokine Signaling Proteins ,Biology ,SH2 domain ,Transfection ,Suppressor of cytokine signalling ,behavioral disciplines and activities ,Leukemia Inhibitory Factor ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,src Homology Domains ,Mice ,Suppressor of Cytokine Signaling 1 Protein ,otorhinolaryngologic diseases ,Animals ,Phosphorylation ,Molecular Biology ,Lymphokines ,General Immunology and Microbiology ,Interleukin-6 ,General Neuroscience ,JNK Mitogen-Activated Protein Kinases ,Proteins ,Cell Differentiation ,Glycoprotein 130 ,Molecular biology ,Growth Inhibitors ,Cell biology ,DNA-Binding Proteins ,Repressor Proteins ,Suppressor of Cytokine Signaling 3 Protein ,Calcium-Calmodulin-Dependent Protein Kinases ,Mutation ,Trans-Activators ,Cytokines ,Tyrosine ,sense organs ,Signal transduction ,Mitogen-Activated Protein Kinases ,Janus kinase ,Carrier Proteins ,Leukemia inhibitory factor ,psychological phenomena and processes ,Signal Transduction ,Transcription Factors ,Research Article - Abstract
SOCS-1 (suppressor of cytokine signaling-1) is a representative of a family of negative regulators of cytokine signaling (SOCS-1 to SOCS-7 and CIS) characterized by a highly conserved C-terminal SOCS box preceded by an SH2 domain. This study comprehensively examined the ability of several SOCS family members to negatively regulate the gp130 signaling pathway. SOCS-1 and SOCS-3 inhibited both interleukin-6 (IL-6)- and leukemia inhibitory factor (LIF)-induced macrophage differentiation of murine monocytic leukemic M1 cells and LIF induction of a Stat3-responsive reporter construct in 293T fibroblasts. Deletion of amino acids 51-78 in the N-terminal region of SOCS-1 prevented inhibition of LIF signaling. The SOCS-1 and SOCS-3 N-terminal regions were functionally interchangeable, but this did not extend to other SOCS family members. Mutation of SH2 domains abrogated the ability of both SOCS-1 and SOCS-3 to inhibit LIF signal transduction. Unlike SOCS-1, SOCS-3 was unable to inhibit JAK kinase activity in vitro, suggesting that SOCS-1 and SOCS-3 act on the JAK-STAT pathway in different ways. Thus, although inhibition of signaling by SOCS-1 and SOCS-3 requires both the SH2 and N-terminal domains, their mechanisms of action appear to be biochemically different.
- Published
- 1999
47. IL-11 Antagonist Inhibits Decidual Transformation Resulting in Pregnancy Failure in Mice
- Author
-
Lois A. Salamonsen, Louis Fabri, Ellen Menkhorst, Felicity Meredith Dunlop, Manuel Baca, Andrew D. Nash, Pierre Scotney, and Evdokia Dimitriadis
- Subjects
Transformation (genetics) ,medicine.medical_specialty ,Pregnancy ,Endocrinology ,Reproductive Medicine ,Internal medicine ,Reproductive biology ,Antagonist ,medicine ,Cell Biology ,General Medicine ,Biology ,medicine.disease - Published
- 2008
- Full Text
- View/download PDF
48. Phage display of a catalytic antibody to optimize affinity for transition-state analog binding
- Author
-
Robert C. Stephenson, Thomas S. Scanlan, James A. Wells, and Manuel Baca
- Subjects
chemistry.chemical_classification ,Multidisciplinary ,Phage display ,biology ,Genetic Vectors ,Mutagenesis (molecular biology technique) ,Antibodies, Catalytic ,Biological Sciences ,Combinatorial chemistry ,Catalysis ,Mice ,Enzyme ,chemistry ,Transition state analog ,Mutagenesis ,biology.protein ,Animals ,Humans ,Bacteriophages ,Binding Sites, Antibody ,Antibody ,Cloning, Molecular ,Panning (camera) ,Hapten ,Haptens - Abstract
Catalytic antibodies have shown great promise for catalyzing a tremendously diverse set of natural and unnatural chemical transformations. However, few catalytic antibodies have efficiencies that approach those of natural enzymes. In principle, random mutagenesis procedures such as phage display could be used to improve the catalytic activities of existing antibodies; however, these studies have been hampered by difficulties in the recombinant expression of antibodies. Here, we have grafted the antigen binding loops from a murine-derived catalytic antibody, 17E8, onto a human antibody framework in an effort to overcome difficulties associated with recombinant expression and phage display of this antibody. “Humanized” 17E8 retained similar catalytic and hapten binding properties as the murine antibody while levels of functional Fab displayed on phage were 200-fold higher than for a murine variable region/human constant region chimeric Fab. This construct was used to prepare combinatorial libraries. Affinity panning of these resulted in the selection of variants with 2- to 8-fold improvements in binding affinity for a phosphonate transition-state analog. Surprisingly, none of the affinity-matured variants was more catalytically active than the parent antibody and some were significantly less active. By contrast, a weaker binding variant was identified with 2-fold greater catalytic activity and incorporation of a single substitution (Tyr-100aH→ Asn) from this variant into the parent antibody led to a 5-fold increase in catalytic efficiency. Thus, phage display methods can be readily used to optimize binding of catalytic antibodies to transition-state analogs, and when used in conjunction with limited screening for catalysis can identify variants with higher catalytic efficiencies.
- Published
- 1997
49. Antibody humanization using monovalent phage display
- Author
-
James A. Wells, Shane J. O'Connor, Leonard G. Presta, and Manuel Baca
- Subjects
Vascular Endothelial Growth Factor A ,Phage display ,Protein Conformation ,Mutant ,Molecular Sequence Data ,Computational biology ,Endothelial Growth Factors ,Biology ,Humanized antibody ,Biochemistry ,Chimera (genetics) ,Immunoglobulin Fab Fragments ,Mice ,Protein structure ,Antigen ,Escherichia coli ,Animals ,Humans ,Bacteriophages ,Amino Acid Sequence ,Binding site ,Cloning, Molecular ,Codon ,Molecular Biology ,Gene Library ,Lymphokines ,Base Sequence ,Vascular Endothelial Growth Factors ,Antibodies, Monoclonal ,Cell Biology ,Molecular biology ,Recombinant Proteins ,Models, Structural ,Oligodeoxyribonucleotides ,Mutagenesis ,biology.protein ,Binding Sites, Antibody ,Antibody ,Immunoglobulin Heavy Chains - Abstract
Antibody humanization often requires the replacement of key residues in the framework regions with corresponding residues from the parent non-human antibody. These changes are in addition to grafting of the antigen-binding loops. Although guided by molecular modeling, assessment of which framework changes are beneficial to antigen binding usually requires the analysis of many different antibody mutants. Here we describe a phage display method for optimizing the framework of humanized antibodies by random mutagenesis of important framework residues. We have applied this method to humanization of the anti-vascular endothelial growth factor murine monoclonal antibody A4.6.1. Affinity panning of a library of humanized A4.6.1 antibody mutants led to the selection of one variant with greater than 125-fold enhanced affinity for antigen relative to the initial humanized antibody with no framework changes. A single additional mutation gave a further 6-fold improvement in binding. The affinity of this variant, 9.3 nM, was only 6-fold weaker than that of a murine/human chimera of A4.6.1. This method provides a general means of rapidly selecting framework mutations that improve the binding of humanized antibodies to their cognate antigens and may prove an attractive alternative to current methods of framework optimization based on cycles of site-directed mutagenesis.
- Published
- 1997
50. Abstract 239: A novel multimeric protein scaffold stimulates apoptotic signaling through TRAILR2
- Author
-
Rosa A. Carrasco, Bob Hollingsworth, Ivan Inigo, David A. Tice, Ching-Ching Leow, Manuel Baca, Luba Grinberg, Herren Wu, Hong Chen, Lin Wang, Jeffery Swers, and Zhan Xiao
- Subjects
Scaffold protein ,Cancer Research ,Programmed cell death ,biology ,Cancer ,Ligand (biochemistry) ,medicine.disease ,Oncology ,Apoptosis ,Immunology ,Cancer cell ,medicine ,Cancer research ,biology.protein ,Receptor ,Caspase - Abstract
TRAILR2 (DR5, TNFRSF10B) is a member of the TNF-receptor superfamily that mediates programmed cell death following stimulation with its ligand. Binding of homotrimeric ligand TRAIL to TRAILR2 induces oligomerization of the receptors and triggers the apoptotic pathway through induction of caspases. Although TRAILR2 is found to be expressed in a number of normal cell types it is routinely found at high levels in many cancer types. Since TRAIL has been shown to preferentially induce apoptosis in cancer cells versus normal cells, several TRAIL receptor agonists including monoclonal antibodies as well as the ligand itself, are currently in clinical development. Here, we describe a novel protein scaffold (Tn3) based on a fibronectin type-III domain from the protein Tenascin that was engineered to specifically agonize TRAILR2. Connecting TRAILR2-binding Tn3 domains together significantly improved the in vitro potency of the molecule in several cancer cell models. For example, a hexameric TRAILR2-binding Tn3 construct has an EC50 of 0.5 pM in a 72 hour cell viability assay with Colo-205 cells. This is approximately 2 logs better than TRAIL. Furthermore, a subset of TRAIL-resistant cancer cell lines derived from multiple tissues of origin is sensitive to the multimeric Tn3. The improved potency over TRAIL was demonstrated in colorectal cancer xenograft models. In addition, tumor regression was also observed at doses as low as 0.3 mg/kg. These results suggest that a multimeric Tn3 protein scaffold targeting TRAILR2 has more potent anti-tumor activity in vitro and in vivo compared than the natural ligand. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 239. doi:1538-7445.AM2012-239
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.