Network


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

Hotspot


Dive into the research topics where Heinrich Haas is active.

Publication


Featured researches published by Heinrich Haas.


Nature | 2016

Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy

Lena M. Kranz; Mustafa Diken; Heinrich Haas; Sebastian Kreiter; Carmen Loquai; Kerstin C. Reuter; Martin Meng; Daniel Fritz; Fulvia Vascotto; Hossam Hefesha; Christian Grunwitz; Mathias Vormehr; Yves Hüsemann; Abderraouf Selmi; Andreas Kuhn; Janina Buck; Evelyna Derhovanessian; Richard Rae; Sebastian Attig; Jan Diekmann; Robert A. Jabulowsky; Sandra Heesch; Jessica C. Hassel; Peter Langguth; Stephan Grabbe; Christoph Huber; Özlem Türeci; Ugur Sahin

Lymphoid organs, in which antigen presenting cells (APCs) are in close proximity to T cells, are the ideal microenvironment for efficient priming and amplification of T-cell responses. However, the systemic delivery of vaccine antigens into dendritic cells (DCs) is hampered by various technical challenges. Here we show that DCs can be targeted precisely and effectively in vivo using intravenously administered RNA-lipoplexes (RNA-LPX) based on well-known lipid carriers by optimally adjusting net charge, without the need for functionalization of particles with molecular ligands. The LPX protects RNA from extracellular ribonucleases and mediates its efficient uptake and expression of the encoded antigen by DC populations and macrophages in various lymphoid compartments. RNA-LPX triggers interferon-α (IFNα) release by plasmacytoid DCs and macrophages. Consequently, DC maturation in situ and inflammatory immune mechanisms reminiscent of those in the early systemic phase of viral infection are activated. We show that RNA-LPX encoding viral or mutant neo-antigens or endogenous self-antigens induce strong effector and memory T-cell responses, and mediate potent IFNα-dependent rejection of progressive tumours. A phase I dose-escalation trial testing RNA-LPX that encode shared tumour antigens is ongoing. In the first three melanoma patients treated at a low-dose level, IFNα and strong antigen-specific T-cell responses were induced, supporting the identified mode of action and potency. As any polypeptide-based antigen can be encoded as RNA, RNA-LPX represent a universally applicable vaccine class for systemic DC targeting and synchronized induction of both highly potent adaptive as well as type-I-IFN-mediated innate immune mechanisms for cancer immunotherapy.


Biophysical Journal | 1995

X-RAY DIFFRACTION OF A PROTEIN CRYSTAL ANCHORED AT THE AIR/WATER INTERFACE

Heinrich Haas; Gerald Brezesinski; Helmuth Möhwald

We report the first successful in situ x-ray diffraction experiment with a 2D protein array at the lipid/water interface and demonstrate that the order can be controlled via lateral pressure or density. A protein (streptavidin) was bound to a monolayer of biotinylated lipid at the air/water interface, and diffraction of the protein layer could be measured to many orders. Compression of the monolayer changed the diffraction pattern drastically, indicating that the protein structure can be strongly influenced by external parameters like lateral pressure or density. From the width of the peaks, we find that aggregates consisting of as few as 100 monomers contribute to the diffraction. This indicates that the structure of even low order aggregates can be studied in situ. Grazing incidence diffraction can become a strong new method to study the crystallization and the interactions between proteins free from artifacts by staining or sample preparation.


Thin Solid Films | 1989

Specific and unspecific binding of concanavalin A at monolayer surfaces

Heinrich Haas; Helmuth Möhwald

Fluorescence and electron microscopic studies with phospholipid monolayers containing concanavalin A and with or without a glycolipid as a specific ligand are presented. It is shown that the (unspecific) protein insertion in the absence of the glycolipid leads to protein patches that may exhibit uniform sizes and that may be due to the existence of long-range electrostatic forces resulting from a difference in the polarization of different protein and lipid areas. By incorporating about 1 mol.% of the glycolipid, specific binding can be effected. This can dominate the unspecific process only in a narrow lateral pressure range between 5 and 15 mN m-1. The data can be qualitatively understood, indicating that control of the interactions requires fine tuning of the head-group arrangement.


Japanese Journal of Applied Physics | 1995

Phospholipid and protein monolayers

Helmuth Möhwald; Hubert Baltes; Michael Schwendler; Christiane A. Helm; Gerald Brezesinski; Heinrich Haas

We report on X-ray reflectivity and diffraction studies using monolayers of phospholipids and of the protein streptavidin specifically bound to monolayers. For phospholipids with the phosphocholine head group attached to the glycerol backbone via (flexible) ethylene oxide spacers, we demonstrate that the lateral interactions can be reduced by increasing of the spacer length. This is reflected in a reduction of the tilt angle of aliphatic tails. Diffraction of the protein layer can be observed in situ. The data reveal that positional order is merely short-ranged and that the structure can be changed reversibly via film compression and expansion.


Chemistry and Physics of Lipids | 1994

Miscibility of lipoteichoic acid in dipalmitoylphosphatidylcholine studied by monofilm investigations and fluorescence microscopy

Thomas Gutberlet; Klaus Milde; Hans Bradaczek; Heinrich Haas; Helmuth Möhwald

The miscibility of the bacterial amphiphile lipoteichoic acid, a constituent of the cytoplasmic membrane of Gram-positive bacteria, in dipalmitoylphosphatidylcholine has been investigated by classic monofilm measurements and fluorescence microscopy at the air-water interface of monofilms obtained by spreading mixtures of both amphiphiles on a water subphase. The isotherms indicated miscibility of both lipids at concentrations up to 30 mol% lipoteichoic acid, whereas at higher concentrations immiscibility was detected. Increasing the lateral pressure over a certain value, lipoteichoic acid is squeezed out of the monofilm. By fluorescence microscopy the influence of lipoteichoic acid on the domain shape of condensed dipalmitoylphosphatidylcholine phases has been studied. The balance between hydrophobic and hydrophilic forces in the mixtures of both amphiphiles is discussed.


Molecular Therapy | 2017

Self-Amplifying RNA Vaccines Give Equivalent Protection against Influenza to mRNA Vaccines but at Much Lower Doses

Annette B. Vogel; Laura Lambert; Ekaterina Kinnear; David Busse; Stephanie Erbar; Kerstin C. Reuter; Lena Wicke; Mario Perkovic; Tim Beissert; Heinrich Haas; Stephen T. Reece; Ugur Sahin; John S. Tregoning

New vaccine platforms are needed to address the time gap between pathogen emergence and vaccine licensure. RNA-based vaccines are an attractive candidate for this role: they are safe, are produced cell free, and can be rapidly generated in response to pathogen emergence. Two RNA vaccine platforms are available: synthetic mRNA molecules encoding only the antigen of interest and self-amplifying RNA (sa-RNA). sa-RNA is virally derived and encodes both the antigen of interest and proteins enabling RNA vaccine replication. Both platforms have been shown to induce an immune response, but it is not clear which approach is optimal. In the current studies, we compared synthetic mRNA and sa-RNA expressing influenza virus hemagglutinin. Both platforms were protective, but equivalent levels of protection were achieved using 1.25 μg sa-RNA compared to 80 μg mRNA (64-fold less material). Having determined that sa-RNA was more effective than mRNA, we tested hemagglutinin from three strains of influenza H1N1, H3N2 (X31), and B (Massachusetts) as sa-RNA vaccines, and all protected against challenge infection. When sa-RNA was combined in a trivalent formulation, it protected against sequential H1N1 and H3N2 challenges. From this we conclude that sa-RNA is a promising platform for vaccines against viral diseases.


Molecular Pharmaceutics | 2017

Incorporation of mRNA in Lamellar Lipid Matrices for Parenteral Administration

Antje Ziller; Sara S. Nogueira; Eva Hühn; Sérgio S. Funari; Gerald Brezesinski; Hermann Hartmann; Ugur Sahin; Heinrich Haas; Peter Langguth

Insertion of high molecular weight messenger RNA (mRNA) into lyotropic lipid phases as model systems for controlled release formulations for the mRNA was investigated. Low fractions of 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) were used as an anchor to load the mRNA into a lamellar lipid matrix. Dispersions of zwitterionic lipid in the aqueous phase in the presence of increasing fractions of mRNA and cationic lipid were prepared, and the molecular organization was investigated as a function of mRNA and cationic lipid fraction. Insertion of both cationic lipid and mRNA was clearly proven from the physicochemical characteristics. The d-spacing of the lipid bilayers, as determined by small-angle X-ray scattering (SAXS) measurements, responded sensitively to the amount of inserted DOTAP and mRNA. A concise model of the insertion of the mRNA in the lipid matrices was derived, indicating that the mRNA was accommodated in the aqueous slab between lipid bilayers. Depending on the DOTAP and mRNA fraction, a different excess of water was present in this slab. Results from further physicochemical characterization, including determination of free and bound mRNA, zeta potential, and calorimetry data, were in line with this assumption. The structure of these concentrated lipid/mRNA preparations was maintained upon dilution. The functionality of the inserted mRNA was proven by cell culture experiments using C2C12 murine myoblast cells with the luciferase-encoding mRNA. The described lipid phases as carriers for the mRNA may be applicable for different routes of local administration, where control of the release kinetics and the form of the released mRNA (bound or free) is required.


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.

Collaboration


Dive into the Heinrich Haas's collaboration.

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

Kerstin C. Reuter

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Janina Buck

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge