Network


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

Hotspot


Dive into the research topics where Thomas Kramps is active.

Publication


Featured researches published by Thomas Kramps.


Nature Biotechnology | 2012

Protective efficacy of in vitro synthesized, specific mRNA vaccines against influenza A virus infection

Benjamin Petsch; Margit Schnee; Annette B. Vogel; Elke Lange; Bernd Hoffmann; Daniel Voss; Thomas Schlake; Andreas Thess; Karl-Josef Kallen; Lothar Stitz; Thomas Kramps

Despite substantial improvements, influenza vaccine production—and availability—remain suboptimal. Influenza vaccines based on mRNA may offer a solution as sequence-matched, clinical-grade material could be produced reliably and rapidly in a scalable process, allowing quick response to the emergence of pandemic strains. Here we show that mRNA vaccines induce balanced, long-lived and protective immunity to influenza A virus infections in even very young and very old mice and that the vaccine remains protective upon thermal stress. This vaccine format elicits B and T cell–dependent protection and targets multiple antigens, including the highly conserved viral nucleoprotein, indicating its usefulness as a cross-protective vaccine. In ferrets and pigs, mRNA vaccines induce immunological correlates of protection and protective effects similar to those of a licensed influenza vaccine in pigs. Thus, mRNA vaccines could address substantial medical need in the area of influenza prophylaxis and the broader realm of anti-infective vaccinology.


PLOS Neglected Tropical Diseases | 2016

An mRNA Vaccine Encoding Rabies Virus Glycoprotein Induces Protection against Lethal Infection in Mice and Correlates of Protection in Adult and Newborn Pigs

Margit Schnee; Annette B. Vogel; Daniel Voss; Benjamin Petsch; Patrick Baumhof; Thomas Kramps; Lothar Stitz

Rabies is a zoonotic infectious disease of the central nervous system (CNS). In unvaccinated or untreated subjects, rabies virus infection causes severe neurological symptoms and is invariably fatal. Despite the long-standing existence of effective vaccines, vaccine availability remains insufficient, with high numbers of fatal infections mostly in developing countries. Nucleic acid based vaccines have proven convincingly as a new technology for the fast development of vaccines against newly emerging pathogens, diseases where no vaccine exists or for replacing already existing vaccines. We used an optimized non-replicating rabies virus glycoprotein (RABV-G) encoding messenger RNA (mRNA) to induce potent neutralizing antibodies (VN titers) in mice and domestic pigs. Functional antibody titers were followed in mice for up to one year and titers remained stable for the entire observation period in all dose groups. T cell analysis revealed the induction of both, specific CD4+ as well as CD8+ T cells by RABV-G mRNA, with the induced CD4+ T cells being higher than those induced by a licensed vaccine. Notably, RABV-G mRNA vaccinated mice were protected against lethal intracerebral challenge infection. Inhibition of viral replication by vaccination was verified by qRT-PCR. Furthermore, we demonstrate that CD4+ T cells are crucial for the generation of neutralizing antibodies. In domestic pigs we were able to induce VN titers that correlate with protection in adult and newborn pigs. This study demonstrates the feasibility of a non-replicating mRNA rabies vaccine in small and large animals and highlights the promises of mRNA vaccines for the prevention of infectious diseases.


Archive | 2012

Messenger RNA Vaccines

Jochen Probst; Mariola Fotin-Mleczek; Thomas Schlake; Andreas Thess; Thomas Kramps; Karl-Josef Kallen

Twenty years after the seminal observation of Wolff et al. that injection of naked RNA and DNA vectors results in protein expression in vivo, messenger RNA (mRNA) vaccines have found entry into clinical development. Through improved vector design, formulation, and delivery, mRNA, initially perceived as unstable and difficult to manipulate, has been developed into a convenient, efficacious, and flexible vaccine platform. Importantly, the same production process can be used to produce a variety of different vaccines, independent of the specifics of particular constructs, which ultimately decreases costs and development time.


Archive | 2017

Introduction to RNA Vaccines

Thomas Kramps; Knut Elbers

RNA vaccines are attractive, because they exhibit characteristics of subunit vaccines and live-attenuated vectors, including flexible production and induction of both humoral and cellular immunity. While human proof-of-concept for RNA vaccines is still pending, the nascent field of RNA therapeutics has already attracted substantial industry and government funding as well as record investments of private venture capital. Most recently, the WHO acknowledged messenger RNA (mRNA) as a new therapeutic class. In this chapter, we briefly review key developments in RNA vaccines and outline the contents of this volume of Methods in Molecular Biology.


PLOS Neglected Tropical Diseases | 2017

A thermostable messenger RNA based vaccine against rabies

Lothar Stitz; Annette B. Vogel; Margit Schnee; Daniel Voss; Susanne Rauch; Thorsten Mutzke; Thomas Ketterer; Thomas Kramps; Benjamin Petsch

Although effective rabies virus vaccines have been existing for decades, each year, rabies virus infections still cause around 50.000 fatalities worldwide. Most of these cases occur in developing countries, where these vaccines are not available. The reasons for this are the prohibitive high costs of cell culture or egg grown rabies virus vaccines and the lack of a functional cold chain in many regions in which rabies virus is endemic. Here, we describe the excellent temperature resistance of a non-replicating mRNA based rabies virus vaccine encoding the rabies virus glycoprotein (RABV-G). Prolonged storage of the vaccine from -80°C to up to +70°C for several months did not impact the protective capacity of the mRNA vaccine. Efficacy after storage was demonstrated by the induction of rabies specific virus neutralizing antibodies and protection in mice against lethal rabies infection. Moreover, storing the vaccine at oscillating temperatures between +4° and +56°C for 20 cycles in order to simulate interruptions of the cold chain during vaccine transport, did not affect the vaccine’s immunogenicity and protective characteristics, indicating that maintenance of a cold chain is not essential for this vaccine.


Journal of Immunology | 2017

A New RNA-Based Adjuvant Enhances Virus-Specific Vaccine Responses by Locally Triggering TLR- and RLH-Dependent Effects

Annett Ziegler; Claudia Soldner; Stefan Lienenklaus; Julia Spanier; Stephanie Trittel; Peggy Riese; Thomas Kramps; Siegfried Weiss; Regina Heidenreich; Edith Jasny; Carlos A. Guzmán; Karl-Josef Kallen; Mariola Fotin-Mleczek; Ulrich Kalinke

Among innovative adjuvants conferring a Th1-shift, RNAdjuvant is a promising candidate. This adjuvant consists of a 547-nt uncapped noncoding ssRNA containing polyU repeats that is stabilized by a cationic carrier peptide. Whereas vaccination of mice with an influenza subunit vaccine induced moderate virus-specific IgG1, vaccination together with RNAdjuvant significantly enhanced this IgG1 and additionally promoted the formation of IgG2b/c, which is indicative of Th1 responses. Furthermore, such sera neutralized influenza virus, whereas this effect was not detected upon vaccination with the subunit vaccine alone. Similarly, upon vaccination with virus-like particles displaying vesicular stomatitis virus G protein, RNAdjuvant promoted the formation of virus-specific IgG2b/c and enhanced neutralizing IgG responses to an extent that mice were protected against lethal virus infection. RNAdjuvant induced dendritic cells to upregulate activation markers and produce IFN-I. Although these effects were strictly TLR7 dependent, RNAdjuvant-mediated augmentation of vaccine responses needed concurrent TLR and RIG-I–like helicase signaling. This was indicated by the absence of the adjuvant effect in vaccinated MyD88−/−Cardif−/− mice, which are devoid of TLR (with the exception of TLR3) and RIG-I–like helicase signaling, whereas in vaccinated MyD88−/− mice the adjuvant effect was reduced. Notably, i.m. RNAdjuvant injection induced local IFN-I responses and did not induce systemic effects, implying good tolerability and a favorable safety profile for RNAdjuvant.


Archive | 2011

Complexation of nucleic acids with disulfide-crosslinked cationic components for transfection and immunostimulation

Patrick Baumhof; Soehnke Voss; Thomas Kramps; Karl-Josef Kallen


Archive | 2013

PHARMACEUTICAL COMPOSITION COMPRISING A POLYMERIC CARRIER CARGO COMPLEX AND AT LEAST ONE PROTEIN OR PEPTIDE ANTIGEN

Patrick Baumhof; Thomas Kramps; Söhnke Voss; Karl-Josef Kallen; Mariola Fotin-Mleczek


Archive | 2012

Vaccination in elderly patients

Karl-Josef Kallen; Thomas Kramps; Margit Schnee; Benjamin Petsch; Lothar Stitz


Archive | 2012

Vaccination in newborns and infants

Karl-Josef Kallen; Thomas Kramps; Margit Schnee; Benjamin Petsch; Lothar Stitz

Collaboration


Dive into the Thomas Kramps's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Benjamin Petsch

Friedrich Loeffler Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Söhnke Voss

University of Tübingen

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