Network


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

Hotspot


Dive into the research topics where Ingelise J. Gordon is active.

Publication


Featured researches published by Ingelise J. Gordon.


Science | 2013

Protection Against Malaria by Intravenous Immunization with a Nonreplicating Sporozoite Vaccine

Robert A. Seder; Lee Jah Chang; Mary E. Enama; Kathryn L. Zephir; Uzma N. Sarwar; Ingelise J. Gordon; LaSonji A. Holman; Eric R. James; Peter F. Billingsley; Anusha Gunasekera; Adam Richman; Sumana Chakravarty; Anita Manoj; Soundarapandian Velmurugan; Minglin Li; Adam Ruben; Tao Li; Abraham G. Eappen; Richard E. Stafford; Sarah Plummer; Cynthia S. Hendel; Laura Novik; Pamela Costner; Floreliz Mendoza; Jamie G. Saunders; Martha Nason; Jason H. Richardson; Jittawadee Murphy; Silas A. Davidson; Thomas L. Richie

Malaria Sporozoite Vaccine Each year, hundreds of millions of people are infected with Plasmodium falciparum, the mosquito-borne parasite that causes malaria. A preventative vaccine is greatly needed. Seder et al. (p. 1359, published online 8 August; see the Perspective by Good) now report the results from a phase I clinical trial where subjects were immunized intravenously with a whole, attenuated sporozoite vaccine. Three of 9 subjects who received four doses and zero of 6 subjects who received five doses of the vaccine went on to develop malaria after controlled malaria infection. Both antibody titers and cellular immune responses correlated positively with the dose of vaccine received, suggesting that both arms of the adaptive immune response may have participated in the observed protection. Intravenous immunization with an attenuated whole malaria sporozoite vaccine protected volunteers in a phase I clinical trial. [Also see Perspective by Good] Consistent, high-level, vaccine-induced protection against human malaria has only been achieved by inoculation of Plasmodium falciparum (Pf) sporozoites (SPZ) by mosquito bites. We report that the PfSPZ Vaccine—composed of attenuated, aseptic, purified, cryopreserved PfSPZ—was safe and wel-tolerated when administered four to six times intravenously (IV) to 40 adults. Zero of six subjects receiving five doses and three of nine subjects receiving four doses of 1.35 × 105 PfSPZ Vaccine and five of six nonvaccinated controls developed malaria after controlled human malaria infection (P = 0.015 in the five-dose group and P = 0.028 for overall, both versus controls). PfSPZ-specific antibody and T cell responses were dose-dependent. These data indicate that there is a dose-dependent immunological threshold for establishing high-level protection against malaria that can be achieved with IV administration of a vaccine that is safe and meets regulatory standards.


Clinical and Vaccine Immunology | 2006

A DNA Vaccine for Ebola Virus Is Safe and Immunogenic in a Phase I Clinical Trial

Julie E. Martin; Nancy J. Sullivan; Mary E. Enama; Ingelise J. Gordon; Mario Roederer; Richard A. Koup; Robert T. Bailer; Bimal K. Chakrabarti; Michael Bailey; Phillip L. Gomez; Charla Andrews; Zoe Moodie; Lin Gu; Judith Stein; Gary J. Nabel; Barney S. Graham

ABSTRACT Ebola viruses represent a class of filoviruses that causes severe hemorrhagic fever with high mortality. Recognized first in 1976 in the Democratic Republic of Congo, outbreaks continue to occur in equatorial Africa. A safe and effective Ebola virus vaccine is needed because of its continued emergence and its potential for use for biodefense. We report the safety and immunogenicity of an Ebola virus vaccine in its first phase I human study. A three-plasmid DNA vaccine encoding the envelope glycoproteins (GP) from the Zaire and Sudan/Gulu species as well as the nucleoprotein was evaluated in a randomized, placebo-controlled, double-blinded, dose escalation study. Healthy adults, ages 18 to 44 years, were randomized to receive three injections of vaccine at 2 mg (n = 5), 4 mg (n = 8), or 8 mg (n = 8) or placebo (n = 6). Immunogenicity was assessed by enzyme-linked immunosorbent assay (ELISA), immunoprecipitation-Western blotting, intracellular cytokine staining (ICS), and enzyme-linked immunospot assay. The vaccine was well-tolerated, with no significant adverse events or coagulation abnormalities. Specific antibody responses to at least one of the three antigens encoded by the vaccine as assessed by ELISA and CD4+ T-cell GP-specific responses as assessed by ICS were detected in 20/20 vaccinees. CD8+ T-cell GP-specific responses were detected by ICS assay in 6/20 vaccinees. This Ebola virus DNA vaccine was safe and immunogenic in humans. Further assessment of the DNA platform alone and in combination with replication-defective adenoviral vector vaccines, in concert with challenge and immune data from nonhuman primates, will facilitate evaluation and potential licensure of an Ebola virus vaccine under the Animal Rule.


The New England Journal of Medicine | 2017

Chimpanzee Adenovirus Vector Ebola Vaccine - Preliminary Report.

Julie E. Ledgerwood; Adam DeZure; Daphne Stanley; Laura Novik; Mary E. Enama; Nina M. Berkowitz; Zonghui Hu; Gyan Joshi; Aurélie Ploquin; Sandra Sitar; Ingelise J. Gordon; Sarah A. Plummer; LaSonji A. Holman; Cynthia S. Hendel; Galina Yamshchikov; François Roman; Alfredo Nicosia; Stefano Colloca; Riccardo Cortese; Robert T. Bailer; Richard M. Schwartz; Mario Roederer; John R. Mascola; Richard A. Koup; Nancy J. Sullivan; Barney S. Graham; Abstr Act

Background The unprecedented 2014 epidemic of Ebola virus disease (EVD) prompted an international response to accelerate the availability of a preventive vaccine. A replication‐defective recombinant chimpanzee adenovirus type 3–vectored ebolavirus vaccine (cAd3‐EBO), encoding the glycoprotein from Zaire and Sudan species, that offers protection in the nonhuman primate model, was rapidly advanced into phase 1 clinical evaluation. Methods We conducted a phase 1, dose‐escalation, open‐label trial of cAd3‐EBO. Twenty healthy adults, in sequentially enrolled groups of 10 each, received vaccination intramuscularly in doses of 2×1010 particle units or 2×1011 particle units. Primary and secondary end points related to safety and immunogenicity were assessed throughout the first 8 weeks after vaccination; in addition, longer‐term vaccine durability was assessed at 48 weeks after vaccination. Results In this small study, no safety concerns were identified; however, transient fever developed within 1 day after vaccination in two participants who had received the 2×1011 particle‐unit dose. Glycoprotein‐specific antibodies were induced in all 20 participants; the titers were of greater magnitude in the group that received the 2×1011 particle‐unit dose than in the group that received the 2×1010 particle‐unit dose (geometric mean titer against the Zaire antigen at week 4, 2037 vs. 331; P=0.001). Glycoprotein‐specific T‐cell responses were more frequent among those who received the 2×1011 particle‐unit dose than among those who received the 2×1010 particle‐unit dose, with a CD4 response in 10 of 10 participants versus 3 of 10 participants (P=0.004) and a CD8 response in 7 of 10 participants versus 2 of 10 participants (P=0.07) at week 4. Assessment of the durability of the antibody response showed that titers remained high at week 48, with the highest titers in those who received the 2×1011 particle‐unit dose. Conclusions Reactogenicity and immune responses to cAd3‐EBO vaccine were dose‐dependent. At the 2×1011 particle‐unit dose, glycoprotein Zaire–specific antibody responses were in the range reported to be associated with vaccine‐induced protective immunity in challenge studies involving nonhuman primates, and responses were sustained to week 48. Phase 2 studies and efficacy trials assessing cAd3‐EBO are in progress. (Funded by the Intramural Research Program of the National Institutes of Health; VRC 207 ClinicalTrials.gov number, NCT02231866.)


Science Translational Medicine | 2015

Virologic effects of broadly neutralizing antibody VRC01 administration during chronic HIV-1 infection

Rebecca M. Lynch; Eli Boritz; Emily E. Coates; Adam DeZure; Patrick Madden; Pamela Costner; Mary E. Enama; Sarah Plummer; LaSonji A. Holman; Cynthia S. Hendel; Ingelise J. Gordon; Joseph P. Casazza; Michelle Conan-Cibotti; Stephen A. Migueles; Randall Tressler; Robert T. Bailer; Adrian B. McDermott; Sandeep Narpala; Sijy O’Dell; Gideon Wolf; Jeffrey D. Lifson; Brandie A. Freemire; Robert J. Gorelick; Janardan P. Pandey; Sarumathi Mohan; Nicolas Chomont; Rémi Fromentin; Tae-Wook Chun; Anthony S. Fauci; Richard M. Schwartz

A single infusion with broadly neutralizing antibody VRC01 resulted in lowered plasma virus load in HIV-1–infected subjects. Passive aggression for HIV Antibodies that neutralize HIV could add to the therapeutic arsenal to prevent and treat disease. Lynch et al. have now tested one such antibody—VRC01—in HIV-infected individuals. Although little difference was observed in viral reservoir in individuals on antiretroviral therapy, plasma viremia was reduced in untreated subjects with a single infusion of VRC01, preferentially suppressing neutralization-sensitive strains. Passive immunization with neutralizing antibodies could therefore aid in viral suppression in HIV-infected individuals. Passive immunization with HIV-1–neutralizing monoclonal antibodies (mAbs) is being considered for prevention and treatment of HIV-1 infection. As therapeutic agents, mAbs could be used to suppress active virus replication, maintain suppression induced by antiretroviral therapy (ART), and/or decrease the size of the persistent virus reservoir. We assessed the impact of VRC01, a potent human mAb targeting the HIV-1 CD4 binding site, on ART-treated and untreated HIV-1–infected subjects. Among six ART-treated individuals with undetectable plasma viremia, two infusions of VRC01 did not reduce the peripheral blood cell–associated virus reservoir measured 4 weeks after the second infusion. In contrast, six of eight ART-untreated, viremic subjects infused with a single dose of VRC01 experienced a 1.1 to 1.8 log10 reduction in plasma viremia. The two subjects with minimal responses to VRC01 were found to have predominantly VRC01-resistant virus before treatment. Notably, two subjects with plasma virus load <1000 copies/ml demonstrated virus suppression to undetectable levels for over 20 days until VRC01 levels declined. Among the remaining four subjects with baseline virus loads between 3000 and 30,000 copies, viremia was only partially suppressed by mAb infusion, and we observed strong selection pressure for the outgrowth of less neutralization-sensitive viruses. In summary, a single infusion of mAb VRC01 significantly decreased plasma viremia and preferentially suppressed neutralization-sensitive virus strains. These data demonstrate the virological effect of this neutralizing antibody and highlight the need for combination strategies to maintain virus suppression.


Lancet Infectious Diseases | 2011

DNA priming and influenza vaccine immunogenicity: two phase 1 open label randomised clinical trials

Julie E. Ledgerwood; Chih-Jen Wei; Zonghui Hu; Ingelise J. Gordon; Mary E. Enama; Cynthia S. Hendel; Patrick M. McTamney; Melissa B. Pearce; Hadi M. Yassine; Jeffrey C. Boyington; Robert T. Bailer; Terrence M. Tumpey; Richard A. Koup; John R. Mascola; Gary J. Nabel; Barney S. Graham

Summary Background Because the general population is largely naive to H5N1 influenza, antibodies generated to H5 allow analysis of novel influenza vaccines independent of background immunity from previous infection. We assessed the safety and immunogenicity of DNA encoding H5 as a priming vaccine to improve antibody responses to inactivated influenza vaccination. Methods In VRC 306 and VRC 310, two sequentially enrolled phase 1, open-label, randomised clinical trials, healthy adults (age 18–60 years) were randomly assigned to receive intramuscular H5 DNA (4 mg) at day 0 or twice, at day 0 and week 4, followed by H5N1 monovalent inactivated vaccine (MIV; 90 μg) at 4 or 24 weeks, and compared with a two-dose regimen of H5N1 MIV with either a 4 or 24 week interval. Antibody responses were assessed by haemagglutination inhibition (HAI), ELISA, neutralisation (ID80), and immunoassays for stem-directed antibodies. T cell responses were assessed by intracellular cytokine staining. After enrolment, investigators and individuals were not masked to group assignment. VRC 306 and VRC 310 are registered with ClinicalTrials.gov, numbers NCT00776711 and NCT01086657, respectively. Findings In VRC 306, 60 individuals were randomly assigned to the four groups (15 in each) and 59 received the vaccinations. In VRC 310, of the 21 individuals enrolled, 20 received the vaccinations (nine received a two-dose regimen of H5N1 MIV and 11 received H5 DNA at day 0 followed by H5N1 MIV at week 24). H5 DNA priming was safe and enhanced H5-specific antibody titres following an H5N1 MIV boost, especially when the interval between DNA prime and MIV boost was extended to 24 weeks. In the two studies, DNA priming with a 24-week MIV boost interval induced protective HAI titres in 21 (81%) of 26 of individuals, with an increase in geometric mean titre (GMT) of more than four times that of individuals given the MIV-MIV regimen at 4 or 24 weeks (GMT 103–206 vs GMT 27–33). Additionally, neutralising antibodies directed to the conserved stem region of H5 were induced by this prime-boost regimen in several individuals. No vaccine-related serious adverse events were recorded. Interpretation DNA priming 24 weeks in advance of influenza vaccine boosting increased the magnitude of protective antibody responses (HAI) and in some cases induced haemagglutinin-stem-specific neutralising antibodies. A DNA-MIV vaccine regimen could enhance the efficacy of H5 or other influenza vaccines and shows that anti-stem antibodies can be elicited by vaccination in man. Funding National Institutes of Health.


The Journal of Infectious Diseases | 2007

A West Nile Virus DNA Vaccine Induces Neutralizing Antibody in Healthy Adults during a Phase 1 Clinical Trial

Julie E. Martin; Theodore C. Pierson; Sarah Hubka; Steve Rucker; Ingelise J. Gordon; Mary E. Enama; Charla A. Andrews; Qing Xu; Brent S. Davis; Martha Nason; Michael P. Fay; Richard A. Koup; Mario Roederer; Robert T. Bailer; Phillip L. Gomez; John R. Mascola; Gwong-Jen J. Chang; Gary J. Nabel; Barney S. Graham

BACKGROUND West Nile virus (WNV) is a mosquito-borne flavivirus that can cause severe meningitis and encephalitis in infected individuals. We report the safety and immunogenicity of a WNV DNA vaccine in its first phase 1 human study. METHODS A single-plasmid DNA vaccine encoding the premembrane and the envelope glycoproteins of the NY99 strain of WNV was evaluated in an open-label study in 15 healthy adults. Twelve subjects completed the 3-dose vaccination schedule, and all subjects completed 32 weeks of evaluation for safety and immunogenicity. The development of a vaccine-induced immune response was assessed by enzyme-linked immunosorbant assay, neutralization assays, intracelluar cytokine staining, and enzyme-linked immunospot assay. RESULTS The vaccine was safe and well tolerated, with no significant adverse events. Vaccine-induced T cell and antibody responses were detected in the majority of subjects. Neutralizing antibody to WNV was detected in all subjects who completed the 3-dose vaccination schedule, at levels shown to be protective in studies of horses, an incidental natural host for WNV. CONCLUSIONS Further assessment of this DNA platform for human immunization against WNV is warranted. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT00106769 .


Clinical and Experimental Immunology | 2015

Safety, pharmacokinetics and neutralization of the broadly neutralizing HIV-1 human monoclonal antibody VRC01 in healthy adults

Julie E. Ledgerwood; Emily E. Coates; Galina Yamshchikov; Jamie G. Saunders; LaSonji A. Holman; Mary E. Enama; Adam DeZure; Rebecca M. Lynch; Ingelise J. Gordon; Sarah A. Plummer; Cynthia S. Hendel; Amarendra Pegu; Michelle Conan-Cibotti; Sandra Sitar; Robert T. Bailer; Sandeep Narpala; Adrian B. McDermott; Mark K. Louder; Sijy O'Dell; Sarumathi Mohan; Janardan P. Pandey; Richard M. Schwartz; Zonghui Hu; Richard A. Koup; Edmund V. Capparelli; John R. Mascola; Barney S. Graham

VRC‐HIVMAB060‐00‐AB (VRC01) is a broadly neutralizing HIV‐1 monoclonal antibody (mAb) isolated from the B cells of an HIV‐infected patient. It is directed against the HIV‐1 CD4 binding site and is capable of potently neutralizing the majority of diverse HIV‐1 strains. This Phase I dose‐escalation study in healthy adults was conducted at the National Institutes of Health (NIH) Clinical Center (Bethesda, MD, USA). Primary objectives were the safety, tolerability and pharmacokinetics (PK) of VRC01 intravenous (i.v.) infusion at 5, 20 or 40 mg/kg, given either once (20 mg/kg) or twice 28 days apart (all doses), and of subcutaneous (s.c.) delivery at 5 mg/kg compared to s.c. placebo given twice, 28 days apart. Cumulatively, 28 subjects received 43 VRC01 and nine received placebo administrations. There were no serious adverse events or dose‐limiting toxicities. Mean 28‐day serum trough concentrations after the first infusion were 35 and 57 μg/ml for groups infused with 20 mg/kg (n = 8) and 40 mg/kg (n = 5) doses, respectively. Mean 28‐day trough concentrations after the second infusion were 56 and 89 μg/ml for the same two doses. Over the 5–40 mg/kg i.v. dose range (n = 18), the clearance was 0·016 l/h and terminal half‐life was 15 days. After infusion VRC01 retained expected neutralizing activity in serum, and anti‐VRC01 antibody responses were not detected. The human monoclonal antibody (mAb) VRC01 was well tolerated when delivered i.v. or s.c. The mAb demonstrated expected half‐life and pharmacokinetics for a human immunoglobulin G. The safety and PK results support and inform VRC01 dosing schedules for planning HIV‐1 prevention efficacy studies.


Nature Medicine | 2016

Protection against malaria at 1 year and immune correlates following PfSPZ vaccination

Andrew S. Ishizuka; Kirsten E. Lyke; Adam DeZure; Andrea A. Berry; Thomas L. Richie; Floreliz Mendoza; Mary E. Enama; Ingelise J. Gordon; Lee-Jah Chang; Uzma N Sarwar; Kathryn L. Zephir; LaSonji A. Holman; Eric R. James; Peter F. Billingsley; Anusha Gunasekera; Sumana Chakravarty; Anita Manoj; Minglin Li; Adam Ruben; Tao Li; Abraham G. Eappen; Richard E. Stafford; Natasha K C; Tooba Murshedkar; Hope DeCederfelt; Sarah Plummer; Cynthia S. Hendel; Laura Novik; Pamela Costner; Jamie G. Saunders

An attenuated Plasmodium falciparum (Pf) sporozoite (SPZ) vaccine, PfSPZ Vaccine, is highly protective against controlled human malaria infection (CHMI) 3 weeks after immunization, but the durability of protection is unknown. We assessed how vaccine dosage, regimen, and route of administration affected durable protection in malaria-naive adults. After four intravenous immunizations with 2.7 × 105 PfSPZ, 6/11 (55%) vaccinated subjects remained without parasitemia following CHMI 21 weeks after immunization. Five non-parasitemic subjects from this dosage group underwent repeat CHMI at 59 weeks, and none developed parasitemia. Although Pf-specific serum antibody levels correlated with protection up to 21–25 weeks after immunization, antibody levels waned substantially by 59 weeks. Pf-specific T cell responses also declined in blood by 59 weeks. To determine whether T cell responses in blood reflected responses in liver, we vaccinated nonhuman primates with PfSPZ Vaccine. Pf-specific interferon-γ-producing CD8 T cells were present at ∼100-fold higher frequencies in liver than in blood. Our findings suggest that PfSPZ Vaccine conferred durable protection to malaria through long-lived tissue-resident T cells and that administration of higher doses may further enhance protection.


Vaccine | 2008

A SARS DNA vaccine induces neutralizing antibody and cellular immune responses in healthy adults in a Phase I clinical trial.

Julie E. Martin; Mark K. Louder; LaSonji A. Holman; Ingelise J. Gordon; Mary E. Enama; Brenda D. Larkin; Charla A. Andrews; Leatrice Vogel; Richard A. Koup; Mario Roederer; Robert T. Bailer; Phillip L. Gomez; Martha Nason; John R. Mascola; Gary J. Nabel; Barney S. Graham

Abstract Background The severe acute respiratory syndrome (SARS) virus is a member of the Coronaviridae (CoV) family that first appeared in the Guangdong Province of China in 2002 and was recognized as an emerging infectious disease in March 2003. Over 8000 cases and 900 deaths occurred during the epidemic. We report the safety and immunogenicity of a SARS DNA vaccine in a Phase I human study. Methods A single-plasmid DNA vaccine encoding the Spike (S) glycoprotein was evaluated in 10 healthy adults. Nine subjects completed the 3 dose vaccination schedule and were evaluated for vaccine safety and immune responses. Immune response was assessed by intracellular cytokine staining (ICS), ELISpot, ELISA, and neutralization assays. Results The vaccine was well tolerated. SARS-CoV-specific antibody was detected by ELISA in 8 of 10 subjects and neutralizing antibody was detected in all subjects who received 3 doses of vaccine. SARS-CoV-specific CD4+ T-cell responses were detected in all vaccinees, and CD8+ T-cell responses in ∼20% of individuals. Conclusions The VRC SARS DNA vaccine was well tolerated and produced cellular immune responses and neutralizing antibody in healthy adults.


The Journal of Infectious Diseases | 2014

DNA Vaccines Encoding Ebolavirus and Marburgvirus Wild Type Glycoproteins are Safe and Immunogenic in a Phase I Clinical Trial

Uzma N. Sarwar; Pamela Costner; Mary E. Enama; Nina M. Berkowitz; Zonghui Hu; Cynthia S. Hendel; Sandra Sitar; Sarah Plummer; Sabue Mulangu; Robert T. Bailer; Richard A. Koup; John R. Mascola; Gary J. Nabel; Nancy J. Sullivan; Barney S. Graham; Julie E. Ledgerwood; Ingelise J. Gordon; LaSonji A. Holman; Floreliz Mendoza; Laura Novik; Jamie G. Saunders; Kathy Zephir; Niraj Desai; Sheryl Young; Joseph P. Casazza; Brenda D. Larkin; Galina Yamshchikov; Olga Vasilenko; Phillip L. Gomez; Charla Andrews

Background Ebolavirus and Marburgvirus cause severe hemorrhagic fever with high mortality and are potential bioterrorism agents. There are no available vaccines or therapeutic agents. Previous clinical trials evaluated transmembrane-deleted and point-mutation Ebolavirus glycoproteins (GPs) in candidate vaccines. Constructs evaluated in this trial encode wild-type (WT) GP from Ebolavirus Zaire and Sudan species and the Marburgvirus Angola strain expressed in a DNA vaccine. Methods The VRC 206 study evaluated the safety and immunogenicity of these DNA vaccines (4 mg administered intramuscularly by Biojector) at weeks 0, 4, and 8, with a homologous boost at or after week 32. Safety evaluations included solicited reactogenicity and coagulation parameters. Primary immune assessment was done by means of GP-specific enzyme-linked immunosorbent assay. Results The vaccines were well tolerated, with no serious adverse events; 80% of subjects had positive enzyme-linked immunosorbent assay results (≥30) at week 12. The fourth DNA vaccination boosted the immune responses. Conclusions The investigational Ebolavirus and Marburgvirus WT GP DNA vaccines were safe, well tolerated, and immunogenic in this phase I study. These results will further inform filovirus vaccine research toward a goal of inducing protective immunity by using WT GP antigens in candidate vaccine regimens. Clinical Trials Registration NCT00605514.

Collaboration


Dive into the Ingelise J. Gordon's collaboration.

Top Co-Authors

Avatar

Mary E. Enama

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Robert T. Bailer

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Barney S. Graham

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

LaSonji A. Holman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Richard A. Koup

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Cynthia S. Hendel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

John R. Mascola

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Julie E. Ledgerwood

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Laura Novik

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jamie G. Saunders

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge