Network


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

Hotspot


Dive into the research topics where Reiner Laus is active.

Publication


Featured researches published by Reiner Laus.


Journal of Clinical Oncology | 2000

Immunotherapy of Hormone-Refractory Prostate Cancer With Antigen-Loaded Dendritic Cells

Eric J. Small; Paige Fratesi; David M. Reese; George Strang; Reiner Laus; Madhusudan V. Peshwa; Frank Valone

PURPOSE Provenge (Dendreon Corp, Seattle, WA) is an immunotherapy product consisting of autologous dendritic cells loaded ex vivo with a recombinant fusion protein consisting of prostatic acid phosphatase (PAP) linked to granulocyte-macrophage colony-stimulating factor. Sequential phase I and phase II trials were performed to determine the safety and efficacy of Provenge and to assess its capacity to break immune tolerance to the normal tissue antigen PAP. PATIENTS AND METHODS All patients had hormone-refractory prostate cancer. Dendritic-cell precursors were harvested by leukapheresis in weeks 0, 4, 8, and 24, loaded ex vivo with antigen for 2 days, and then infused intravenously over 30 minutes. Phase I patients received increasing doses of Provenge, and phase II patients received all the Provenge that could be prepared from a leukapheresis product. RESULTS Patients tolerated treatment well. Fever, the most common adverse event, occurred after 15 infusions (14.7%). All patients developed immune responses to the recombinant fusion protein used to prepare Provenge, and 38% developed immune responses to PAP. Three patients had a more than 50% decline in prostate-specific antigen (PSA) level, and another three patients had 25% to 49% decreases in PSA. The time to disease progression correlated with development of an immune response to PAP and with the dose of dendritic cells received. CONCLUSION Provenge is a novel immunotherapy agent that is safe and breaks tolerance to the tissue antigen PAP. Preliminary evidence for clinical efficacy warrants further exploration.


The Prostate | 1998

Induction of prostate tumor‐specific CD8+ cytotoxic T‐lymphocytes in vitro using antigen‐presenting cells pulsed with prostatic acid phosphatase peptide

Madhusudan V. Peshwa; Jia Dong Shi; Curtis L. Ruegg; Reiner Laus; Wim C.A. van Schooten

Most strategies in cancer immunotherapy are aimed at the induction of a strong cellular immune response against the tumor. Particularly, CD8+ T lymphocytes have been proven in multiple animal models to be critical for the eradication of solid tumors.


Cancer Research | 2011

Exosome Targeting of Tumor Antigens Expressed by Cancer Vaccines Can Improve Antigen Immunogenicity and Therapeutic Efficacy

Ryan Rountree; Stefanie Mandl; James M. Nachtwey; Katie Dalpozzo; Lisa Do; John R. Lombardo; Peter L. Schoonmaker; Kay Brinkmann; Ulrike Dirmeier; Reiner Laus; Alain Delcayre

MVA-BN-PRO (BN ImmunoTherapeutics) is a candidate immunotherapy product for the treatment of prostate cancer. It encodes 2 tumor-associated antigens, prostate-specific antigen (PSA), and prostatic acid phosphatase (PAP), and is derived from the highly attenuated modified vaccinia Ankara (MVA) virus stock known as MVA-BN. Past work has shown that the immunogenicity of antigens can be improved by targeting their localization to exosomes, which are small, 50- to 100-nm diameter vesicles secreted by most cell types. Exosome targeting is achieved by fusing the antigen to the C1C2 domain of the lactadherin protein. To test whether exosome targeting would improve the immunogenicity of PSA and PAP, 2 additional versions of MVA-BN-PRO were produced, targeting either PSA (MVA-BN-PSA-C1C2) or PAP (MVA-BN-PAP-C1C2) to exosomes, while leaving the second transgene untargeted. Treatment of mice with MVA-BN-PAP-C1C2 led to a striking increase in the immune response against PAP. Anti-PAP antibody titers developed more rapidly and reached levels that were 10- to 100-fold higher than those for mice treated with MVA-BN-PRO. Furthermore, treatment with MVA-BN-PAP-C1C2 increased the frequency of PAP-specific T cells 5-fold compared with mice treated with MVA-BN-PRO. These improvements translated into a greater frequency of tumor rejection in a PAP-expressing solid tumor model. Likewise, treatment with MVA-BN-PSA-C1C2 increased the antigenicity of PSA compared with treatment with MVA-BN-PRO and resulted in a trend of improved antitumor efficacy in a PSA-expressing tumor model. These experiments confirm that targeting antigen localization to exosomes is a viable approach for improving the therapeutic potential of MVA-BN-PRO in humans.


Cancer Letters | 1998

Selective apoptosis of neoplastic cells by an hla-dr specific monoclonal antibody

Damir Vidovic; Reiner Laus

The human major histocompatibility complex (MHC) class II molecule-specific monoclonal antibody (mAb) 8D1 can induce apoptosis of tumor cells expressing HLA-DR molecules on their surface. This effect is associated with a cross-linking of HLA-DR, since monovalent Fab fragments of 8D1 cannot mediate cytotoxicity unless they are anchored to a solid support. Anti-neoplastic activity of 8D1 is highly selective, i.e. the mAb affects neither the viability nor the function of non-malignant HLA-DR+ cells. These findings raise the possibility of a selective antibody-based anti-tumor therapy of class II positive blood cell neoplasm.


Nature Biotechnology | 2000

Enhanced major histocompatibility complex class I-dependent presentation of antigens modified with cationic and fusogenic peptides.

Reiner Laus; Thomas Graddis; Itzhak Hakim; Damir Vidovic

Soluble extracellular protein antigens are notoriously poor stimulators of CD8+ cytotoxic T-lymphocyte (CTL) responses, largely because these antigens have inefficient access to an endogenous cytosolic pathway of the major histocompatibility complex (MHC) class I–dependent antigen presentation. Here, we present a strategy that facilitates antigen penetration into the cytosol of antigen-presenting cells (APC) by addition to the antigen of charge-modifying peptide sequences. As a result of this intervention, the charge modification enhances antigen uptake into APC by counteracting the repulsive cell surface charge, and then endosomal membranes are disrupted with a subsequent release of antigen into the cytosol. This technology significantly improves MHC class I–dependent antigen presentation to CTL, enabling a more efficient generation of specific CTL immunity in vivo. The strategy described here has potential for use in developing efficient vaccines for antigen-specific immunotherapy of human malignancies.


Cancer Immunology, Immunotherapy | 2012

Immunotherapy with MVA-BN®-HER2 induces HER-2-specific Th1 immunity and alters the intratumoral balance of effector and regulatory T cells.

Stefanie Mandl; Ryan Rountree; Katie Dalpozzo; Lisa Do; John R. Lombardo; Peter L. Schoonmaker; Ulrike Dirmeier; Robin Steigerwald; Thierry Giffon; Reiner Laus; Alain Delcayre

MVA-BN®-HER2 is a new candidate immunotherapy designed for the treatment of HER-2-positive breast cancer. Here, we demonstrate that a single treatment with MVA-BN®-HER2 exerts potent anti-tumor efficacy in a murine model of experimental pulmonary metastasis. This anti-tumor efficacy occurred despite a strong tumor-mediated immunosuppressive environment characterized by a high frequency of regulatory T cells (Treg) in the lungs of tumor-bearing mice. Immunogenicity studies showed that treatment with MVA-BN®-HER2 induced strongly Th1-dominated HER-2-specific antibody and T-cell responses. MVA-BN®-HER2-induced anti-tumor activity was characterized by an increased infiltration of lungs with highly activated, HER-2-specific, CD8+CD11c+ T cells accompanied by a decrease in the frequency of Treg cells in the lung, resulting in a significantly increased ratio of effector T cells to Treg cells. In contrast, administration of HER2 protein formulated in Complete Freund’s Adjuvant (CFA) induced a strongly Th2-biased immune response to HER-2. However, this did not lead to significant infiltration of the tumor-bearing lungs by CD8+ T cells or the decrease in the frequency of Treg cells nor did it result in anti-tumor efficacy. In vivo depletion of CD8+ cells confirmed that CD8 T cells were required for the anti-tumor activity of MVA-BN®-HER2. Furthermore, depletion of CD4+ or CD25+ cells demonstrated that tumor-induced Treg cells promoted tumor growth and that CD4 effector cells also contribute to MVA-BN®-HER2-mediated anti-tumor efficacy. Taken together, our data demonstrate that treatment with MVA-BN®-HER2 controls tumor growth through mechanisms including the induction of Th1-biased HER-2-specific immune responses and the control of tumor-mediated immunosuppression.


International Journal of Cancer | 2002

Antitumor vaccination with HER-2-derived recombinant antigens

Damir Vidovic; Thomas Graddis; Feng Chen; Paul Slagle; Michael Diegel; Lara Porter Stepan; Reiner Laus

Certain types of malignant tumors overexpress HER‐2, a transmembrane glycoprotein of the class I receptor tyrosine kinase erbB family. To develop an effective HER‐2 vaccine for the selective immunotherapy of these malignancies, we have genetically engineered fusion proteins containing portions of extra‐ and intracellular HER‐2 domains. Activated dendritic cells (DC) cocultured with these novel antigens (Ag) could induce potent responses of Ag‐specific T‐cell lines in vitro and a protection against HER‐2‐expressing tumor in vivo. The protective capabilities of HER‐2‐derived fusion proteins correlated with the efficiency of their presentation to Ag‐specific T‐cell hybridomas. The most effective Ag contained GM‐CSF, the presence of which facilitated their internalization by antigen‐presenting cells (APC) in a receptor‐mediated manner.


Human Immunology | 2003

Specific stimulation of MHC-transgenic mouse T-cell hybridomas with xenogeneic APC.

Damir Vidovic; Thomas Graddis; Lara Porter Stepan; Dennis M. Zaller; Reiner Laus

From the recombinant human leukocyte antigen (HLA)-DR1/H2-E(k) major histocompatibility complex (MHC) class II-transgenic mice, we have generated two CD4(+) T-cell hybridomas specific for peptides which were derived from human prostatic acid phosphatase (PAP) complexed to the human class II molecule HLA-DR1. Both hybridomas strongly react to PAP-pulsed antigen-presenting cells (APC) from transgenic mice. Interestingly, these hybridomas also responded to PAP antigen presented by HLA-DR1-positive human APC. The species-mismatched T-cell stimulation occurs despite the biologic discordance in participating accessory molecules, which are required for the optimal T-cell-APC interaction. Our results demonstrate various degrees of functional interaction between coreceptors, costimulatory molecules, and integrins, which are expressed on the surface of T-cell hybridomas and heterologous APC.


Scandinavian Journal of Immunology | 2003

Major Histocompatibility Complex Class I-Restricted Presentation of Protein Antigens without Prior Intracellular Processing

Michael Diegel; Feng Chen; Reiner Laus; Thomas Graddis; Damis Vidovic

Proteins in their native form are incapable of stimulating antigen (Ag)‐specific T cells, which can only recognize major histocompatibility complex (MHC)‐bound peptides that have been generated by intracellular processing within antigen‐presenting cells (APCs). Here, we show that APCs can trigger MHC class I‐restricted T‐cell responses after presenting proteins without conventional intracellular processing, provided the immunostimulatory MHC class I‐binding peptide sequence is incorporated at the carboxy‐terminal position. Such MHC‐bound proteins do not stimulate T cells directly, because the contact between MHC/peptide complex and its cognate ligand is sterically hindered by the amino‐terminal bulk of the protein. Removal of the latter via an extracellular Ag proteolysis by the T‐cell‐ and/or APC‐derived enzymes is required for effective T‐cell stimulation. Our data challenge the established concept that only small peptides can bind to the MHC class I molecules.


Cancer Research | 2013

Abstract A21: Active immunotherapy with PROSTVAC® demonstrates potent antitumor efficacy in a mouse model of prostate cancer.

Ryan Rountree; Stefanie Mandl; Joseph Cote; Tracy dela Cruz; Thierry Giffon; Evan Gordon; Susan Foy; John R. Lombardo; Erica Trent; Reiner Laus; Alain Delcayre

BN ImmunoTherapeutics (BNIT) specializes in developing novel active immunotherapies for cancer. These therapies use recombinant poxviruses engineered to express tumor-associated antigens (TAAs), with the intent of generating effective immune responses against the patients9 cancer. PROSTVAC® is a candidate product for the treatment of prostate cancer for which a global Phase III clinical trial (PROSPECT) was recently initiated. This product is composed of two different viral vectors derived from a recombinant vaccinia virus (PROSTVAC™-V) and a recombinant fowlpox virus (PROSTVAC™-F). Both vectors contain transgenes encoding prostate-specific antigen (PSA) and a triad of costimulatory molecules (B7-1, ICAM-1, and LFA-3), designated as TRICOM™. Patients are immunized using a prime-boost strategy consisting of an initial treatment with PROSTVAC™-V followed by repeated boosting with PROSTVAC™-F to maximize the immune responses against the PSA tumor-antigen. Here we show preclinical data characterizing PROSTVAC® activity in mice. Treatment with either PROSTVAC™-V or PROSTVAC™-F induced PSA-specific antibody and T cell responses; however, PSA-specific responses were further increased by the prime/boost strategy, particularly with respect to the frequency of responding CD8 T cells. These CD8 T cells produced IFN-gamma and degranulated in an antigen-specific manner. Furthermore, PROSTVAC® treatment resulted in strong efficacy in a mouse model of prostate cancer. In this model, treatment with PROSTVAC resulted in anti-tumor efficacy accompanied by a Th1-biased response against PSA. In contrast, growth of tumors in control mice induced only non-protective PSA-specific responses with strong Th2 bias. Overall, these animal studies help define the activity and mechanism of action of PROSTVAC® which is currently being evaluated in the clinic. R.B. Rountree and S.J. Mandl contributed equally to this work. Citation Format: Ryan B. Rountree, Stefanie J. Mandl, Joseph Cote, Tracy dela Cruz, Thierry Giffon, Evan Gordon, Susan P. Foy, John R. Lombardo, Erica Trent, Reiner Laus, Alain Delcayre. Active immunotherapy with PROSTVAC ® demonstrates potent antitumor efficacy in a mouse model of prostate cancer. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology: Multidisciplinary Science Driving Basic and Clinical Advances; Dec 2-5, 2012; Miami, FL. Philadelphia (PA): AACR; Cancer Res 2013;73(1 Suppl):Abstract nr A21.

Collaboration


Dive into the Reiner Laus's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge