Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Anna Z. Wec is active.

Publication


Featured researches published by Anna Z. Wec.


Cell | 2017

Antibodies from a Human Survivor Define Sites of Vulnerability for Broad Protection against Ebolaviruses

Anna Z. Wec; Andrew S. Herbert; Charles D. Murin; Elisabeth K. Nyakatura; Dafna M. Abelson; J. Maximilian Fels; Shihua He; Rebekah M. James; Marc Antoine de La Vega; Wenjun Zhu; Russell R. Bakken; Eileen Goodwin; Hannah L. Turner; Rohit K. Jangra; Larry Zeitlin; Xiangguo Qiu; Jonathan R. Lai; Laura M. Walker; Andrew B. Ward; John M. Dye; Kartik Chandran; Zachary A. Bornholdt

Experimental monoclonal antibody (mAb) therapies have shown promise for treatment of lethal Ebola virus (EBOV) infections, but their species-specific recognition of the viral glycoprotein (GP) has limited their use against other divergent ebolaviruses associated with human disease. Here, we mined the human immune response to natural EBOV infection and identified mAbs with exceptionally potent pan-ebolavirus neutralizing activity and protective efficacy against three virulent ebolaviruses. These mAbs recognize an inter-protomer epitope in the GP fusion loop, a critical and conserved element of the viral membrane fusion machinery, and neutralize viral entry by targeting a proteolytically primed, fusion-competent GP intermediate (GPCL) generated in host cell endosomes. Only a few somatic hypermutations are required for broad antiviral activity, and germline-approximating variants display enhanced GPCL recognition, suggesting that such antibodies could be elicited more efficiently with suitably optimized GP immunogens. Our findings inform the development of both broadly effective immunotherapeutics and vaccines against filoviruses.


Cell Reports | 2016

Antibody Treatment of Ebola and Sudan Virus Infection via a Uniquely Exposed Epitope within the Glycoprotein Receptor-Binding Site

Katie A. Howell; Xiangguo Qiu; Jennifer M. Brannan; Christopher Bryan; Edgar Davidson; Frederick W. Holtsberg; Anna Z. Wec; Sergey Shulenin; Julia E. Biggins; Robin Douglas; Sven Enterlein; Hannah L. Turner; Jesper Pallesen; Charles D. Murin; Shihua He; Andrea Kroeker; Hong Vu; Andrew S. Herbert; Marnie L. Fusco; Elisabeth K. Nyakatura; Jonathan R. Lai; Zhen Yong Keck; Steven K. H. Foung; Erica Ollmann Saphire; Larry Zeitlin; Andrew B. Ward; Kartik Chandran; Benjamin J. Doranz; Gary P. Kobinger; John M. Dye

Summary Previous efforts to identify cross-neutralizing antibodies to the receptor-binding site (RBS) of ebolavirus glycoproteins have been unsuccessful, largely because the RBS is occluded on the viral surface. We report a monoclonal antibody (FVM04) that targets a uniquely exposed epitope within the RBS; cross-neutralizes Ebola (EBOV), Sudan (SUDV), and, to a lesser extent, Bundibugyo viruses; and shows protection against EBOV and SUDV in mice and guinea pigs. The antibody cocktail ZMapp™ is remarkably effective against EBOV (Zaire) but does not cross-neutralize other ebolaviruses. By replacing one of the ZMapp™ components with FVM04, we retained the anti-EBOV efficacy while extending the breadth of protection to SUDV, thereby generating a cross-protective antibody cocktail. In addition, we report several mutations at the base of the ebolavirus glycoprotein that enhance the binding of FVM04 and other cross-reactive antibodies. These findings have important implications for pan-ebolavirus vaccine development and defining broadly protective antibody cocktails.


Science | 2016

A "Trojan horse" bispecific antibody strategy for broad protection against ebolaviruses.

Anna Z. Wec; Elisabeth K. Nyakatura; Andrew S. Herbert; Katie A. Howell; Frederick W. Holtsberg; Russell R. Bakken; Eva Mittler; John R. Christin; Sergey Shulenin; Rohit K. Jangra; Sushma Bharrhan; Ana I. Kuehne; Zachary A. Bornholdt; Andrew I. Flyak; Erica Ollmann Saphire; James E. Crowe; M. Javad Aman; John M. Dye; Jonathan R. Lai; Kartik Chandran

Treating Ebola with a Trojan horse The recent major Ebola virus outbreak in West Africa high-lighted the need for effective therapeutics against this and other filoviruses. Neutralizing ebolaviruses with antibodies is a challenge because the viruses bind their entry receptor, NPC1, inside the cell within endosomes rather than on the cell surface. Furthermore, enzymes in endosomes cleave the Ebola virus surface glycoprotein (GP) to reveal its receptor binding site. Wec et al. now report a bispecific antibody strategy targeting all known ebolaviruses that overcomes this problem (see the Perspective by Labrijn and Parren). They coupled an antibody specific for a conserved, surface-exposed epitope of GP to antibodies that recognize either NPC1 or the NPC1 binding site on GP. Treating mice therapeutically with these antibodies allowed them to survive otherwise lethal ebolavirus infection. Science, this issue p. 350; see also p. 284 Bispecific antibodies show therapeutic efficacy against ebolaviruses in mice. There is an urgent need for monoclonal antibody (mAb) therapies that broadly protect against Ebola virus and other filoviruses. The conserved, essential interaction between the filovirus glycoprotein, GP, and its entry receptor Niemann-Pick C1 (NPC1) provides an attractive target for such mAbs but is shielded by multiple mechanisms, including physical sequestration in late endosomes. Here, we describe a bispecific-antibody strategy to target this interaction, in which mAbs specific for NPC1 or the GP receptor–binding site are coupled to a mAb against a conserved, surface-exposed GP epitope. Bispecific antibodies, but not parent mAbs, neutralized all known ebolaviruses by coopting viral particles themselves for endosomal delivery and conferred postexposure protection against multiple ebolaviruses in mice. Such “Trojan horse” bispecific antibodies have potential as broad antifilovirus immunotherapeutics.


mAbs | 2017

Generation and characterization of protective antibodies to Marburg virus

Jeffrey W. Froude; Thibaut Pelat; Sebastian Miethe; Samantha E. Zak; Anna Z. Wec; Kartik Chandran; Jennifer M. Brannan; Russell R. Bakken; Michael Hust; Philippe Thullier; John M. Dye

ABSTRACT Marburg virus (MARV) and Ebola virus (EBOV) have been a source of epidemics and outbreaks for several decades. We present here the generation and characterization of the first protective antibodies specific for wild-type MARV. Non-human primates (NHP), cynomolgus macaques, were immunized with viral-replicon particles expressing the glycoproteins (GP) of MARV (Ci67 isolate). An antibody fragment (single-chain variable fragment, scFv) phage display library was built after four immunogen injections, and screened against the GP1-649 of MARV. Sequencing of 192 selected clones identified 18 clones with distinct VH and VL sequences. Four of these recombinant antibodies (R4A1, R4B11, R4G2, and R3F6) were produced in the scFv-Fc format for in vivo studies. Mice that were challenged with wild-type Marburg virus (Ci67 isolate) receiving 100 µg of scFv-Fc on days −1, 1 and 3 demonstrated protective efficacies ranging from 75–100%. The amino-acid sequences of the scFv-Fcs are similar to those of their human germline counterparts, sharing an identity ranging between 68 and 100% to human germline immunoglobulin. These results demonstrate for the first time that recombinant antibodies offer protection against wild-type MARV, and suggest they may be promising candidates for further therapeutic development especially due to their human homology.


Journal of Biological Chemistry | 2018

Design and evaluation of bi- and trispecific antibodies targeting multiple filovirus glycoproteins

Elisabeth K. Nyakatura; Samantha E. Zak; Anna Z. Wec; Daniel Hofmann; Sergey Shulenin; Russell R. Bakken; M. Javad Aman; Kartik Chandran; John M. Dye; Jonathan R. Lai

Filoviruses (family Filoviridae) include five ebolaviruses and Marburg virus. These pathogens cause a rapidly progressing and severe viral disease with high mortality rates (generally 30–90%). Outbreaks of filovirus disease are sporadic and, until recently, were limited to less than 500 cases. However, the 2013–2016 epidemic in western Africa, caused by Ebola virus (EBOV), illustrated the potential of filovirus outbreaks to escalate to a much larger scale (over 28,000 suspected cases). mAbs against the envelope glycoprotein represent a promising therapeutic platform for managing filovirus infections. However, mAbs that exhibit neutralization or protective properties against multiple filoviruses are rare. Here we examined a panel of engineered bi- and trispecific antibodies, in which variable domains of mAbs that target epitopes from multiple filoviruses were combined, for their capacity to neutralize viral infection across filovirus species. We found that bispecific combinations targeting EBOV and Sudan virus (another ebolavirus), provide potent cross-neutralization and protection in mice. Furthermore, trispecific combinations, targeting EBOV, Sudan virus, and Marburg virus, exhibited strong neutralization potential against all three viruses. These results provide important insights into multispecific antibody engineering against filoviruses and will inform future immunotherapeutic discoveries.


Cell Host & Microbe | 2018

A Role for Fc Function in Therapeutic Monoclonal Antibody-Mediated Protection against Ebola Virus

Bronwyn M. Gunn; Wen-Han Yu; Marcus Karim; Jennifer M. Brannan; Andrew S. Herbert; Anna Z. Wec; Peter Halfmann; Marnie L. Fusco; Sharon L. Schendel; Karthik Gangavarapu; Tyler B. Krause; Xiangguo Qiu; Shinhua He; Jishnu Das; Todd J. Suscovich; Jonathan R. Lai; Kartik Chandran; Larry Zeitlin; James E. Crowe; Douglas A. Lauffenburger; Yoshihiro Kawaoka; Gary P. Kobinger; Kristian G. Andersen; John M. Dye; Erica Ollmann Saphire; Galit Alter

The recent Ebola virus (EBOV) epidemic highlighted the need for effective vaccines and therapeutics to limit and prevent outbreaks. Host antibodies against EBOV are critical for controlling disease, and recombinant monoclonal antibodies (mAbs) can protect from infection. However, antibodies mediate an array of antiviral functions including neutralization as well as engagement of Fc-domain receptors on immune cells, resulting in phagocytosis or NK cell-mediated killing of infected cells. Thus, to understand the antibody features mediating EBOV protection, we examined specific Fc features associated with protection using a library of EBOV-specific mAbs. Neutralization was strongly associated with therapeutic protection against EBOV. However, several neutralizing mAbs failed to protect, while several non-neutralizing or weakly neutralizing mAbs could protect. Antibody-mediated effector functions, including phagocytosis and NK cell activation, were associated with protection, particularly for antibodies with moderate neutralizing activity. This framework identifies functional correlates that can inform therapeutic and vaccine design strategies against EBOV and other pathogens.


bioRxiv | 2018

Structural Basis of Broad Ebolavirus Neutralization by a Human Survivor Antibody

Brandyn R. West; Anna Z. Wec; Crystal L. Moyer; Marnie L. Fusco; Philipp A Illinykh; Kai Huang; Rebekah M. James; Andrew P. Herbert; Sean Hui; Ariel S. Wirchnianski; Eileen Goodwin; M. Javad Aman; Laura M. Walker; John M. Dye; Alexander Bukreyev; Kartik Chandran; Erica Ollmann Saphire

The structural features that govern broad-spectrum activity of broadly neutralizing, anti-ebolavirus antibodies (Abs) are currently unknown. Here we describe the first structure of a broadly neutralizing human Ab, ADI-15946, in complex with cleaved Ebola virus glycoprotein (EBOV GPCL). We find that ADI-15946 employs structural mimicry of a conserved interaction between the GP core and the glycan cap β17-β18 loop to inhibit infection. Both endosomal proteolysis of EBOV GP and binding of monoclonal Ab (mAb) FVM09 displace this loop, increase exposure of ADI-15946’s conserved epitope and potentiate neutralization. Our work also illuminated the determinants of ADI-15946’s reduced activity against Sudan virus (SUDV), and enabled rational, structure-guided engineering to enhance binding and neutralization against SUDV while retaining the parental breadth of activity. One Sentence Summary The first crystal structure of a broadly active antibody against surface glycoproteins of ebolaviruses identifies a highly conserved epitope beneath the glycan cap and highlights the molecular requirements for broad ebolavirus neutralization.


bioRxiv | 2018

Human antibody cocktail deploys multiple functions to confer pan-ebolavirus protection

Anna Z. Wec; Zachary A. Bornholdt; Shihua He; Andrew S. Herbert; Eileen Goodwin; Ariel S. Wirchnianski; Bronwyn M. Gunn; Zirui Zhang; Wenjun Zhu; Guodong Liu; Dafna M. Abelson; Crystal L. Moyer; Rohit K. Jangra; Rebekah M. James; Russell R. Bakken; Natasha Bohorova; Ognian Bohorov; Do Han Kim; Michael Pauly; Jesus Velasco; Robert H Bortz; Kevin J. Whaley; Tracey Goldstein; Simon J. Anthony; Galit Alter; Laura M. Walker; John M. Dye; Larry Zeitlin; Xiangguo Qiu; Kartik Chandran

During the unprecedented 2013–2016 Ebola virus disease (EVD) epidemic in Western Africa and in its aftermath, the passive administration of monoclonal antibodies (mAbs) emerged as a promising treatment approach1–7. However, all antibody-based therapeutics currently in advanced development are specific for a single member of the Ebolavirus genus, Ebola virus (EBOV), and ineffective against divergent outbreak-causing ebolaviruses, including Bundibugyo virus (BDBV) and Sudan virus (SUDV)2,3,5,7. Here we advance MBP134, a cocktail of two broadly neutralizing human mAbs targeting the filovirus surface glycoprotein, GP, as a candidate pan-ebolavirus therapeutic. One component of this cocktail is a pan-ebolavirus neutralizing mAb, ADI-15878, isolated from a human EVD survivor8,9. The second, ADI-23774, was derived by affinity maturation of a human mAb8,9 via yeast display to enhance its potency against SUDV. MBP134 afforded exceptionally potent pan-ebolavirus neutralization in vitro and demonstrated greater protective efficacy than ADI-15878 alone in the guinea pig model of lethal EBOV challenge. A second-generation cocktail, MBP134AF, engineered to effectively harness natural killer (NK) cells afforded additional, unprecedented improvements in protective efficacy against EBOV and SUDV in guinea pigs relative to both its precursor and to any mAbs or mAb cocktails tested previously. MBP134AF is a best-in-class mAb cocktail suitable for evaluation as a pan-ebolavirus therapeutic in nonhuman primates.


bioRxiv | 2018

Two-antibody pan-ebolavirus cocktail confers broad therapeutic protection in ferrets and nonhuman primates

Zachary A. Bornholdt; Andrew S. Herbert; Chad E. Mire; Shihua He; Robert W. Cross; Anna Z. Wec; Dafna M. Abelson; Joan B. Geisbert; Rebekah M. James; Niaz Rahim; Wenjun Zhu; Viktoriya Borisevich; Logan Banadyga; Bronwyn M. Gunn; Krystle N. Agans; Eileen Goodwin; Kevin Tierney; William S. Shestowsky; Ognian Bohorov; Natasha Bohorova; Jesus Velasco; Eric Ailor; Do Han Kim; Michael Pauly; Kevin J. Whaley; Galit Alter; Laura M. Walker; Kartik Chandran; Larry Zeitlin; Xiangguo Qiu

All available experimental vaccines and immunotherapeutics1,2 against Ebola virus (EBOV), including rVSV-ZEBOV3 and ZMappTM4, lack activity against other ebolaviruses associated with human disease outbreaks. This year, two separate outbreaks of EBOV in the Democratic Republic of Congo underscored the unpredictable nature of ebolavirus reemergence in a region that has historically experienced outbreaks of the divergent ebolaviruses Sudan virus (SUDV) and Bundibugyo virus (BDBV)5. Here we show that MBP134AF, a pan-ebolavirus therapeutic comprising two broadly neutralizing human antibodies (bNAbs)6,7(see companion manuscript, Wec et al.) could protect against lethal EBOV, SUDV, and BDBV infection in ferrets and nonhuman primates (NHPs). MBP134AF not only not only establishes a viable therapeutic countermeasure to outbreaks caused by antigenically diverse ebolaviruses but also affords unprecedented effectiveness and potency—a single 25-mg/kg dose was fully protective in NHPs. This best-in-class antibody cocktail is the culmination of an intensive collaboration spanning academia, industry and government in response to the 2013-2016 EBOV epidemic6,7 and provides a translational research model for the rapid development of immunotherapeutics targeting emerging infectious diseases.


Cell | 2017

Immunization-Elicited Broadly Protective Antibody Reveals Ebolavirus Fusion Loop as a Site of Vulnerability

Xuelian Zhao; Katie A. Howell; Shihua He; Jennifer M. Brannan; Anna Z. Wec; Edgar Davidson; Hannah L. Turner; Chi I. Chiang; Lin Lei; J. Maximilian Fels; Hong Vu; Sergey Shulenin; Ashley N. Turonis; Ana I. Kuehne; Guodong Liu; Mi Ta; Yimeng Wang; Christopher Sundling; Yongli Xiao; Jennifer S. Spence; Benjamin J. Doranz; Frederick W. Holtsberg; Andrew B. Ward; Kartik Chandran; John M. Dye; Xiangguo Qiu; Yuxing Li; M. Javad Aman

Collaboration


Dive into the Anna Z. Wec's collaboration.

Top Co-Authors

Avatar

Kartik Chandran

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

John M. Dye

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar

Andrew S. Herbert

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar

Jonathan R. Lai

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Shihua He

Public Health Agency of Canada

View shared research outputs
Top Co-Authors

Avatar

Xiangguo Qiu

Public Health Agency of Canada

View shared research outputs
Top Co-Authors

Avatar

Elisabeth K. Nyakatura

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hannah L. Turner

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

M. Javad Aman

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Researchain Logo
Decentralizing Knowledge