Network


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

Hotspot


Dive into the research topics where Oezlem Tuereci is active.

Publication


Featured researches published by Oezlem Tuereci.


Cancer Research | 2017

Abstract CT034: A first-in-human phase I/II clinical trial assessing novel mRNA-lipoplex nanoparticles for potent melanoma immunotherapy

Robert A. Jabulowsky; Carmen Loquai; Jochen Utikal; Jessica C. Hassel; Roland Kaufmann; Evelyna Derhovanessian; Mustafa Diken; Lena M. Kranz; Heinrich Haas; Sebastian Attig; Christine Anft; Janina Buck; Jan Diekmann; Daniel Fritz; Kerstin Hartmann; Alexandra Kemmer-Brueck; Klaus Kuehlcke; Andreas Kuhn; Peter Langguth; Ulrich Luxemburger; Martin Meng; Richard Rae; Fatih Sari; Doreen Schwarck-Kokarakis; Malte Stein; Stephan Grabbe; Sebastian Kreiter; Oezlem Tuereci; Christoph Huber; Ugur Sahin

Therapeutic vaccination with tumor antigen-encoding RNAs by local administration is currently being successfully employed in various clinical trials. Advancing from local to more efficient systemic targeting of antigen-presenting cells (APCs), we have developed pioneering RNA-lipoplex (RNA(LIP)) immunotherapeutics for intravenous application based on the employment of well-known lipid carriers without the need for functionalization of particles with molecular ligands. The novel RNA(LIP) formulation has been engineered to preserve RNA integrity after intravenous injection and physicochemically optimized for efficient uptake and expression of the encoded antigen by APCs in various lymphoid compartments, resulting in the synchronized induction of both potent adaptive as well as type-I-IFN-mediated innate immune responses. The first-in-human phase I/II dose escalation Lipo-MERIT trial (NCT02410733) assesses the safety, tolerability, and biological efficacy of the innovative RNA(LIP) immunotherapy in four study centers in Germany. This is the first example of a clinically applicable and systemic RNA-based cancer vaccine. Following selective antigen stratification on routinely collected tumor samples, eligible patients with malignant melanoma are treated with increasing doses of the tetravalent Lipo-MERIT vaccine - a fixed set of four RNA(LIP) products, each encoding one shared melanoma-associated antigen, i.e. NY-ESO-1, tyrosinase, MAGE-A3, and TPTE, that are administered successively within one treatment cycle. Accompanying correlative biomarker studies and concerted immunological assessments evaluate the pharmacodynamic activity and immunogenicity upon multiple vaccination cycles with the Lipo-MERIT vaccine. As of January 2017, 15 patients have been treated within five dose escalation cohorts thoroughly guided by an independent data safety and monitoring board. Multiple dosing with the Lipo-MERIT vaccine was generally well-tolerated and no dose-limiting toxicities (DLTs) were observed so far. Further patient enrollment is continuing. Detailed information on the ongoing trial, the recruitment and treatment status as well as preliminary data on the assessment of vaccine-induced immune responses from the first patients treated will be presented. Citation Format: Robert A. Jabulowsky, Carmen Loquai, Jochen Utikal, Jessica Hassel, Roland Kaufmann, Evelyna Derhovanessian, Mustafa Diken, Lena M. Kranz, Heinrich Haas, Sebastian Attig, Christine Anft, Janina Buck, Jan Diekmann, Daniel Fritz, Kerstin Hartmann, Alexandra Kemmer-Brueck, Klaus Kuehlcke, Andreas N. Kuhn, Peter Langguth, Ulrich Luxemburger, Martin Meng, Richard Rae, Fatih Sari, Doreen Schwarck-Kokarakis, Malte Stein, Stephan Grabbe, Sebastian Kreiter, Oezlem Tuereci, Christoph Huber, Ugur Sahin. A first-in-human phase I/II clinical trial assessing novel mRNA-lipoplex nanoparticles for potent melanoma immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr CT034. doi:10.1158/1538-7445.AM2017-CT034


Cancer immunology research | 2016

Abstract B041: A novel nanoparticular formulated tetravalent RNA cancer vaccine for treatment of patients with malignant melanoma

Robert A. Jabulowsky; Carmen Loquai; Mustafa Diken; Lena M. Kranz; Heinrich Haas; Sebastian Attig; Cedrik M. Britten; Janina Buck; Evelyna Derhovanessian; Jan Diekmann; Isaac Esparza; Daniel Fritz; Yves Huesemann; Veronika Jahndel; Klaus Kuehlcke; Andreas Kuhn; Peter Langguth; Ulrich Luxemburger; Martin Meng; Felicitas Mueller; Kerstin C. Reuter; Doreen Schwarck; Kristina Spiess; Meike Witt; Jessica C. Hassel; Jochen Utikal; Roland Kaufmann; Marc Schrott; Sebastian Kreiter; Oezlem Tuereci

Immunotherapeutic approaches have evolved as promising and valid alternatives to available conventional cancer treatments. Amongst others, vaccination with tumor antigen-encoding RNAs by local administration is currently successfully employed in various clinical trials. To allow for a more efficient targeting of antigen-presenting cells (APCs) we have developed a novel RNA immunotherapeutic for systemic application based on a fixed set of four liposome complexed RNA drug products (RNA(LIP)) each encoding one shared melanoma-associated antigen. Similar to other liposomal drugs, the four injectable RNA(LIP) products constituting the investigational medicinal product will be prepared individually in a straight-forward manner directly prior to use from three components, namely solutions containing RNA drug product, NaCl diluent, and liposome excipient, that are provided as a kit. The novel lipoplex formulation was engineered (i) to protect RNA from degradation by plasma RNases and (ii) to enable directed in vivo targeting of APCs in lymphoid compartments, thus (iii) allowing for intravenous administration of multiple RNA products advancing from local to systemic targeting of APCs. The improved selective delivery of the RNA(LIP) products into APCs has further been shown to lead to an enhanced induction of vaccine-induced T-cell responses. Extensive pharmacological characterization of the RNA(LIP) platform revealed that upon cellular uptake the encoded antigens will be translated into proteins that will be rapidly processed into peptide fragments, which after presentation by MHC class I and II molecules on the surface of APCs induce tumor antigen-specific CD8+ and CD4+ T-cell responses that spread systemically. These vaccine-induced T cells have been shown to specifically recognize and kill antigen-positive tumor cells eliciting potent anti-tumoral activity in vivo. The potent vaccination effects are additionally enhanced by further immunomodulatory effects based on the transient release of pro-inflammatory cytokines such as IFN-α, IP-10, and IL-6 due to binding of the administered RNA drug products to Toll-like receptors (TLRs). The clinical translation of this pioneering therapeutic concept is currently being realized in a multi-center, first-in-human phase I trial in patients with malignant melanoma. Main objectives of the clinical trial are to study safety, tolerability, and immunogenicity of this innovative immunotherapy approach. The novel lipoplex formulation, RNA(LIP) mechanism of action, study design and clinical workflow, as well as recruitment and treatment status of the ongoing clinical trial will be presented. Citation Format: Robert A. Jabulowsky, Carmen Loquai, Mustafa Diken, Lena M. Kranz, Heinrich Haas, Sebastian Attig, Cedrik M. Britten, Janina Buck, Evelyna Derhovanessian, Jan Diekmann, Isaac Esparza, Daniel Fritz, Yves Huesemann, Veronika Jahndel, Klaus Kuehlcke, Andreas N. Kuhn, Peter Langguth, Ulrich Luxemburger, Martin Meng, Felicitas Mueller, Kerstin C. Reuter, Doreen Schwarck, Kristina Spiess, Meike Witt, Jessica C. Hassel, Jochen Utikal, Roland Kaufmann, Marc Schrott, Sebastian Kreiter, Oezlem Tuereci, Christoph Huber, Ugur Sahin. A novel nanoparticular formulated tetravalent RNA cancer vaccine for treatment of patients with malignant melanoma. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr B041.


Cancer Research | 2016

Abstract CT032: A first-in-human phase I/II clinical trial assessing novel mRNA-lipoplex nanoparticles for potent cancer immunotherapy in patients with malignant melanoma

Robert A. Jabulowsky; Carmen Loquai; Mustafa Diken; Lena M. Kranz; Heinrich Haas; Sebastian Attig; Nicole Bidmon; Janina Buck; Evelyna Derhovanessian; Jan Diekmann; Daniel Fritz; Veronika Jahndel; Alexandra Kemmer-Brueck; Klaus Kuehlcke; Andreas Kuhn; Peter Langguth; Ulrich Luxemburger; Martin Meng; Felicitas Mueller; Richard Rae; Fatih Sari; Doreen Schwarck-Kokarakis; Christine Seck; Kristina Spieß; Meike Witt; Jessica C. Hassel; Jochen Utikal; Roland Kaufmann; Sebastian Kreiter; Christoph Huber

Immunotherapeutic approaches have evolved as promising and valid alternatives to available conventional cancer treatments. Amongst others, vaccination with tumor antigen-encoding RNAs by local administration is currently successfully employed in various clinical trials. To allow for a more efficient targeting of antigen-presenting cells (APCs) and to overcome potential technical challenges associated with local administration, we have developed a novel RNA immunotherapeutic for systemic application based on a fixed set of four liposome complexed RNA drug products (RNA(LIP)), each encoding one shared melanoma-associated antigen. The novel RNA(LIP) formulation was engineered (i) to protect RNA from degradation by plasma RNases and (ii) to enable directed in vivo targeting of APCs in lymphoid compartments, thus (iii) allowing for intravenous administration of multiple RNA products advancing from local to systemic targeting of APCs. Here, RNA(LIP) products trigger a Toll-like receptor (TLR)-mediated Interferon-α (IFN-α) release from plasmacytoid dendritic cells (DCs) and macrophages stimulating DC maturation and hence inducing innate immune mechanisms as well as potent vaccine antigen-specific immune responses. Notably, BioNTech RNA Pharmaceuticals′ RNA(LIP) formulation is a universally applicable potent novel vaccine class for intravenous APC targeting and the induction of potent synchronized adaptive and type-I interferon-mediated innate immune responses for cancer immunotherapy. Similar to other liposomal drugs, the ready-to-use RNA(LIP) products are prepared individually in a straight-forward manner directly prior to use from three components, namely solutions containing RNA drug product, NaCl diluent, and liposome excipient, that are provided as a kit. A multi-center phase I/II trial to clinically validate this pioneering RNA(LIP) formulation for the treatment of malignant melanoma was initiated in 2015 (NCT02410733). The objective of the clinical trial is to study the feasibility, safety, tolerability, immunogenicity and evaluate potential clinical activity of the RNA(LIP) immunotherapy concept. Detailed information on the ongoing trial, the recruitment and treatment status as well as data on the assessment of vaccine-induced immune responses will be presented. Citation Format: Robert A. Jabulowsky, Carmen Loquai, Mustafa Diken, Lena M. Kranz, Heinrich Haas, Sebastian Attig, Nicole Bidmon, Janina Buck, Evelyna Derhovanessian, Jan Diekmann, Daniel Fritz, Veronika Jahndel, Alexandra Kemmer-Brueck, Klaus Kuehlcke, Andreas N. Kuhn, Peter Langguth, Ulrich Luxemburger, Martin Meng, Felicitas Mueller, Richard Rae, Fatih Sari, Doreen Schwarck-Kokarakis, Christine Seck, Kristina Spies, Meike Witt, Jessica C. Hassel, Jochen Utikal, Roland Kaufmann, Sebastian Kreiter, Christoph Huber, Oezlem Tuereci, Ugur Sahin. A first-in-human phase I/II clinical trial assessing novel mRNA-lipoplex nanoparticles for potent cancer immunotherapy in patients with malignant melanoma. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr CT032.


Cancer Research | 2014

Abstract 2903: IMAB362, a novel first-in-class monoclonal antibody for treatment of pancreatic cancer

Oezlem Tuereci; Stefan Woell; Stefan Jacobs; Rita Mitnacht-Kraus; Ugur Sahin

Pancreatic ductal adenocarcinoma (PDAC), the most frequent subtype (>80%) of pancreatic cancer (PC) is characterized by a generally lethal progress within a short period of time after primary diagnosis. Despite high efforts, the treatment options are very limited and mainly of palliative nature. Therefore, we investigated whether IMAB362 might represent a potential treatment option in this patient population. IMAB362 is a highly potent and tumor-cell selective therapeutic antibody which is currently in clinical development in gastro-esophageal cancer (in phase II and IIb trials). IMAB362 is directed against the tight junction molecule CLDN18.2, a proliferation-promoting transmembrane protein, which is maintained during malignant transformation. Our previous studies demonstrated, that CLDN18.2 is expressed in >55% of patients with PDAC and frequently in metastases at considerable levels. To evaluate the anti-tumor activity of IMAB362 we performed in vitro and in vivo experiments on CLDN18.2 expressing PC cell lines. IMAB362 exerts its anti-tumor function by antibody-dependant cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). ADCC was assessed in various cancer cell lines and peripheral blood mononuclear cells (PBMCs) at an E:T-ratio of 40:1 in the presence of IMAB362. The efficacy ranged from 67.1 ± 6.8 to 92.6 ± 5.5%. EC50 was determined between 23.4 ± 12.3 ng/ml and 5.7 ± 4.4 µg/ml. IMAB3629s capacity to induce CDC was analyzed by incubation of various tumor cell lines with healthy human serum as source for complement. We observed a dose-dependent lysis with EC50 values ranging from 0.3 - 2.6 µg/ml. In vivo studies in PC tumor bearing nude mice showed that administration of repeated doses of IMAB362 tends to result in inhibition of tumor growth and prolonged survival. Also, the anti-tumor effect of IMAB362 was strictly dependent on the expression of its target CLDN18.2. These results demonstrate that IMAB362 has antitumoral activity in vitro and in vivo against PC cell lines and PC xenografts. Most promising, the efficacy was due to IMAB3629s single agent activity only. Combinations with established chemotherapies (e.g. 5-FU, gemcitabine) might deliver a strong rationale for clinical testing of the highly selective monoclonal antibody drug in patients with PDAC. Results from further in vivo testing of IMAB362 combinations with established chemotherapies will be presented at the meeting. Citation Format: Oezlem Tuereci, Stefan Woell, Stefan Jacobs, Rita Mitnacht-Kraus, Ugur Sahin. IMAB362, a novel first-in-class monoclonal antibody for treatment of pancreatic cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2903. doi:10.1158/1538-7445.AM2014-2903


Cancer immunology research | 2016

Abstract B040: Oncolytic measles virus for tumor vaccination

Stefan Hutzler; Stephanie Erbar; Robert A. Jabulowsky; Tim Beissert; Jan R.H. Hanauer; Oezlem Tuereci; Rita Mitnacht-Kraus; Sebastian Kreiter; Mustafa Diken; Cedrik M. Britten; Ugur Sahin; Michael D. Muehlebach

The concept of tumor vaccination is based on the induction of robust immune responses against tumor-associated antigens (TAA) selectively expressed within the tumor. Whereas in the case of foreign antigens an immune response can be induced quite easily, induction of appropriate immunity against self-antigens needs potent antigen presentation to break immune tolerance. Tumor vaccines may be further improved by utilizing the properties of oncolytic viruses (OVs), that selectively replicate within and destroy tumor tissue and may further enhance the vaccination effects by supplying danger signals in situ and stimulating the immune system. Immunogenic OVs additionally delivering a specific TAA by in situ antigen presentation can break immmune tolerance, induce a specifically redirected immune response, and thereby may increase therapeutic efficacy. Attenuated measles virus (MV) derived from a vaccine strain is currently tested as oncolytic virotherapeutic in clinical trials. Recombinant MVs additionally reveal excellent vaccine characteristics per se, inducing potent and long lasting immune responses against endogenous and foreign antigens, if the latter are additionally expressed by the recombinant virus. Therefore, we develop and aim to validate novel prototypic replicating MV for simultaneous oncolysis and in situ tumor vaccination. For that purpose, a well characterized tumor-confined TAA of the claudin family is used as vaccination target antigen (GNTP01). Different antigen formats or epitopes of GNTP01 were cloned into the MV genome and recombinant viruses were generated, which express those epitopes in different amounts as expected due to positional effects of the additional transcription units either behind the P (post P) or the H (post H) gene cassettes of Moraten vaccine strain-derived MVvac2 genome, presented on in situ produced particles or not. Based on these data, selected viruses expressing different antigen formats have been chosen for further characterization. The selected viruses were amplified, and unimpaired growth and genetic stability of the inserted TAA were demonstrated. To test their immunogenic properties, MV-susceptible IFNAR-/--CD46Ge mice have been immunized with the different viruses, and humoral as well as cellular immune responses are currently analyzed. In parallel, an immune competent, syngeneic tumor model has been established by employment of transgenic C57/BL6-derived tumor cell lines expressing the target antigen as well as MV-receptors. For three different transgenic tumor cell lines antigen expression and tumorigenicity in MV-susceptible animals has been confirmed. Such tumor-bearing animals will be used for analysis of induced immune responses, anti-tumoral activity, and therapeutic efficacy after immunization with selected viruses that have been shown to induce significant humoral or cellular immune responses. Once the proof-of-concept has been established this innovative oncolytic virus-based approach holds the potential for a novel therapeutic technology platform that synergistically combines the direct tumor-lytic properties of recombinant MVs with the therapeutic induction of potent immune responses depending on the patient´s TAA expression profile. Citation Format: Stefan Hutzler, Stephanie Erbar, Robert A. Jabulowsky, Tim Beissert, Jan Hanauer, Oezlem Tuereci, Rita Mitnacht-Kraus, Sebastian Kreiter, Mustafa Diken, Cedrik M. Britten, Ugur Sahin, Michael D. Muehlebach. Oncolytic measles virus for tumor vaccination. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr B040.


Cancer Research | 2016

Abstract CT022: IVAC® MUTANOME - A first-in-human phase I clinical trial targeting individual mutant neoantigens for the treatment of melanoma

Matthias Miller; Carmen Loquai; Björn-Philipp Kloke; Sebastian Attig; Nicole Bidmon; Stefanie Bolte; Valesca Bukur; Evelyna Derhovanessian; Jan Diekmann; Angela Filbry; Sandra Heesch; Christoph Hoeller; Klaus Kuehlcke; David Langer; Martin Loewer; Felicitas Mueller; Inga Ortseifer; Burkhard Otte; Anna Paruzynski; Richard Rae; Barbara Schroers; Christine Seck; Kristina Spiess; Arbel D. Tadmor; Isabel Vogler; Mathias Vormehr; Alexandra Kemmer-Brueck; Andreas Kuhn; Ulrich Luxemburger; Sebastian Kreiter

One of the hallmarks of cancer is the inherent instability of the genome leading to multiple genomic alterations and epigenetic changes that ultimately drive carcinogenesis. These processes lead to a unique molecular profile of every given tumor and to substantial intratumoral heterogeneity of cancer tissues. Recently, a series of independent reports revealed that pre-formed neoantigen specific T-cell responses are of crucial relevance for the clinical efficacy of immune checkpoint inhibitors. However, spontaneous immune recognition of neoantigens seems to be a rare event with only less than 1% of mutations inducing a T-cell response in the tumor-bearing patient. Accordingly, only patients with a high burden of mutations profit from currently approved therapies. To overcome this restriction, the IVAC® MUTANOME-project harnesses the individual patient9s mutation profile by manufacturing highly potent neoantigen-coding RNA vaccines. To this end, the individual mutation repertoire is identified by next-generation-sequencing, potentially immunogenic mutations are selected and incorporated into a poly-epitopic RNA vaccine that is tailored to activate and expand the individual patient9s neoantigen-specific CD4+ and CD8+ T cells. A phase I study to test this novel concept of an active individualized cancer vaccine for the treatment of malignant melanoma was initiated in 2013 (NCT02035956). Notably, BioNTech RNA Pharmaceutical9s IVAC® MUTANOME trial is the first-in-human trial that introduces a fully personalized RNA vaccine for the treatment of malignant melanoma. The objective of this clinical trial is to study the feasibility, safety, tolerability, immunogenicity and the potential clinical activity of the IVAC® MUTANOME approach. The recruitment of a patient into the trial triggers the multi-step IVAC® MUTANOME process covering (i) the receipt and processing of tumor and blood sample specimens, (ii) the identification, prioritization and confirmation of mutations, (iii) testing of pre-existing immunity against identified tumor mutations, (iv) the selection of mutant neoantigen sequences as vaccine targets, (v) design, production of a DNA lead structure, (vi) GMP manufacturing and release of the patient-specific mRNA, (vii) shipment to the clinical trial site and (viii) the administration of the IMP to patients. Detailed information on the trial, the recruitment and treatment status as well as data on the assessment of vaccine induced immune responses will be presented. Citation Format: Matthias Miller, Carmen Loquai, Bjorn-Philipp Kloke, Sebastian Attig, Nicole Bidmon, Stefanie Bolte, Valesca Bukur, Evelyna Derhovanessian, Jan Diekmann, Angela Filbry, Sandra Heesch, Christoph Hoeller, Klaus Kuehlcke, David Langer, Martin Loewer, Felicitas Mueller, Inga Ortseifer, Burkhard Otte, Anna Paruzynski, Richard Rae, Barbara Schroers, Christine Seck, Kristina Spiess, Arbel D. Tadmor, Isabel Vogler, Mathias Vormehr, Alexandra Kemmer-Brueck, Andreas N. Kuhn, Ulrich Luxemburger, Sebastian Kreiter, Jochen Utikal, Stephan Grabbe, Oezlem Tuereci, Ugur Sahin. IVAC® MUTANOME - A first-in-human phase I clinical trial targeting individual mutant neoantigens for the treatment of melanoma. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr CT022.


Nature Biotechnology | 1999

Generation of tissue-specific and promiscuous HLA ligand databases using DNA microarrays and virtual HLA class II matrices

Tiziana Sturniolo; Elisa Bono; Jiayi Ding; Laura Raddrizzani; Oezlem Tuereci; Ugur Sahin; Michael Braxenthaler; Fabio Gallazzi; Maria Pia Protti; Francesco Sinigaglia; Juergen Hammer


Archive | 2018

MONOCLONAL ANTI-GT468 ANTIBODIES FOR TREATMENT OF CANCER

Michael Koslowski; Ugur Sahin; Oezlem Tuereci; Rita Mitnacht Kraus


Archive | 2013

MONOCLONAL ANTIBODY FOR TREATMENT OF CANCER

Ugur Sahin; ウグル サヒン; Michael Koslowski; ミヒャエル コスロフスキー; Oezlem Tuereci; エズレム テューレヒ


Archive | 2012

Gene product differentially expressed in tumor and use thereof

Oezlem Tuereci; エツレム・テューレツィ; Ugur Sahin; ウグール・サヒン; Michael Koslowski; ミヒャエル コスロヴスキー; Stefan Fritz; ステファン フリッツ; Harald-Gerhard Geppert; ハラルド−ゲルハルド ゲッペルト

Collaboration


Dive into the Oezlem Tuereci's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andreas Kuhn

University of Hohenheim

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge