93 results on '"Toni Weinschenk"'
Search Results
2. RNA editing derived epitopes function as cancer antigens to elicit immune responses
- Author
-
Minying Zhang, Jens Fritsche, Jason Roszik, Leila J. Williams, Xinxin Peng, Yulun Chiu, Chih-Chiang Tsou, Franziska Hoffgaard, Valentina Goldfinger, Oliver Schoor, Amjad Talukder, Marie A. Forget, Cara Haymaker, Chantale Bernatchez, Leng Han, Yiu-Huen Tsang, Kathleen Kong, Xiaoyan Xu, Kenneth L. Scott, Harpreet Singh-Jasuja, Greg Lizee, Han Liang, Toni Weinschenk, Gordon B. Mills, and Patrick Hwu
- Subjects
Science - Abstract
RNA editing is a biological process that creates sequence variation. Here the authors show that peptides generated as a consequence of RNA editing are naturally presented by human leukocyte antigen (HLA) and serve as antigens to elicit anti-tumour immune responses.
- Published
- 2018
- Full Text
- View/download PDF
3. Antigenic expression and spontaneous immune responses support the use of a selected peptide set from the IMA950 glioblastoma vaccine for immunotherapy of grade II and III glioma
- Author
-
Valérie Dutoit, Denis Migliorini, Giulia Ranzanici, Eliana Marinari, Valérie Widmer, Johannes Alexander Lobrinus, Shahan Momjian, Joseph Costello, Paul R. Walker, Hideho Okada, Toni Weinschenk, Christel Herold-Mende, and Pierre-Yves Dietrich
- Subjects
grade ii glioma ,grade iii glioma ,immunotherapy ,peptide vaccine ,ima950 ,spontaneous t cell responses ,astrocytoma ,oligodendroglioma ,ependymoma ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Gliomas are lethal brain tumors that resist standard therapeutic approaches. Immunotherapy is a promising alternative strategy mostly developed in the context of glioblastoma. However, there is a need for implementing immunotherapy for grade II/III gliomas, as these are the most common CNS tumors in young adults with a high propensity for recurrence, making them lethal despite current treatments. We recently identified HLA-A2-restricted tumor-associated antigens by peptide elution from glioblastoma and formulated a multipeptide vaccine (IMA950) evaluated in phase I/II clinical trials with promising results. Here, we investigated expression of the IMA950 antigens in patients with grade II/III astrocytoma, oligodendroglioma or ependymoma, at the mRNA, protein and peptide levels. We report that the BCAN, CSPG4, IGF2BP3, PTPRZ1 and TNC proteins are significantly over-expressed at the mRNA (n = 159) and protein (n = 36) levels in grade II/III glioma patients as compared to non-tumor samples (IGF2BP3 being absent from oligodendroglioma). Most importantly, we detected spontaneous antigen-specific T cell responses to one or more of the IMA950 antigens in 100% and 71% of grade II and grade III patients, respectively (27 patients tested). These patients displayed T cell responses of better quality (higher frequency, broader epitope targeting) than patients with glioblastoma. Detection of spontaneous T cell responses to the IMA950 antigens shows that these antigens are relevant for tumor targeting, which will be best achieved by combination with CD4 epitopes such as the IDH1R132H peptide. Altogether, we provide the rationale for using a selective set of IMA950 peptides for vaccination of patients with grade II/III glioma.
- Published
- 2018
- Full Text
- View/download PDF
4. Immune Tolerance to Tumor Antigens Occurs in a Specialized Environment of the Spleen
- Author
-
Stefano Ugel, Elisa Peranzoni, Giacomo Desantis, Mariacristina Chioda, Steffen Walter, Toni Weinschenk, Jordi C. Ochando, Anna Cabrelle, Susanna Mandruzzato, and Vincenzo Bronte
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Peripheral tolerance to tumor antigens (Ags) is a major hurdle for antitumor immunity. Draining lymph nodes are considered the privileged sites for Ag presentation to T cells and for the onset of peripheral tolerance. Here, we show that the spleen is fundamentally important for tumor-induced tolerance. Splenectomy restores lymphocyte function and induces tumor regression when coupled with immunotherapy. Splenic CD11b+Gr-1intLy6Chi cells, mostly comprising proliferating CCR2+-inflammatory monocytes with features of myeloid progenitors, expand in the marginal zone of the spleen. Here, they alter the normal tissue cytoarchitecture and closely associate with memory CD8+ T cells, cross-presenting tumor Ags and causing their tolerization. Because of its high proliferative potential, this myeloid cell subset is also susceptible to low-dose chemotherapy, which can be exploited as an adjuvant to passive immunotherapy. CCL2 serum levels in cancer patients are directly related to the accumulation of immature myeloid cells and are predictive for overall survival in patients who develop a multipeptide response to cancer vaccines.
- Published
- 2012
- Full Text
- View/download PDF
5. Moderate degradation does not preclude microarray analysis of small amounts of RNA
- Author
-
Oliver Schoor, Toni Weinschenk, Jörg Hennenlotter, Stefan Corvin, Arnulf Stenzl, Hans-Georg Rammensee, and Stefan Stevanović
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Gene expression analysis by microarrays using small amounts of RNA is becoming more and more popular against the background of advances and increasing importance of small-sample acquisition methods like laser microdissection techniques. The quality of RNA preparations from such samples constitutes a frequent issue in this context. The aim of this study was to assess the impact of different extents of RNA degradation on the expression profile of the samples. We induced RNA degradation in human tumor and healthy tissue samples by endogeneous ribonucleases. Next, we amplified 20 ng total RNA degraded to different extents by two rounds of in vitro transcription and analyzed them using Affymetrix oligonucleotide microarrays. Expression differences for some genes were independently confirmed by real-time quantitative PCR. Our results suggest that gene expression profiles obtained from partially degraded RNA samples with still visible ribosomal bands exhibit a high degree of similarity compared to intact samples and that RNA samples of suboptimal quality might therefore still lead to meaningful results if used carefully.
- Published
- 2003
- Full Text
- View/download PDF
6. Feasibility and Safety of Personalized, Multi-Target, Adoptive Cell Therapy (IMA101): First-In-Human Clinical Trial in Patients with Advanced Metastatic Cancer
- Author
-
Apostolia M. Tsimberidou, Kerstin Guenther, Borje S. Andersson, Regina Mendrzyk, Amir Alpert, Claudia Wagner, Anna Nowak, Katrin Aslan, Arun Satelli, Fabian Richter, Sabrina Kuttruff-Coqui, Oliver Schoor, Jens Fritsche, Zoe Coughlin, Ali S. Mohamed, Kerry Sieger, Becky Norris, Rita Ort, Jennifer Beck, Henry Hiep. Vo, Franziska Hoffgaard, Manuel Ruh, Linus Backert, Ignacio I. Wistuba, David Fuhrmann, Nuhad K. Ibrahim, Van Karlyle. Morris, Bryan K. Kee, Daniel M. Halperin, Graciela M. Nogueras-González, Partow Kebriaei, Elizabeth J. Shpall, David Vining, Patrick Hwu, Harpreet Singh-Jasuja, Carsten Reinhardt, Cedrik M. Britten, Norbert Hilf, Toni Weinschenk, Dominik Maurer, and Steffen Walter
- Subjects
Cancer Research ,Immunology - Abstract
IMA101 is an actively personalized, multi-targeted adoptive cell therapy (ACT), whereby autologous T cells are directed against multiple novel defined peptide-HLA (pHLA) cancer targets. HLA-A*02:01-positive patients with relapsed/refractory solid tumors expressing ≥1 of 8 pre-defined targets underwent leukapheresis. Endogenous T cells specific for up to 4 targets were primed and expanded in vitro. Patients received lymphodepletion (fludarabine, cyclophosphamide), followed by T-cell infusion and low-dose interleukin-2 (Cohort 1). Patients in Cohort 2 received atezolizumab for up to 1 year (NCT02876510). Overall, 214 patients were screened, 15 received lymphodepletion (13 women, 2 men; median age, 44 years), and 14 were treated with T-cell products. IMA101 treatment was feasible and well tolerated. The most common adverse events were cytokine release syndrome (Grade 1, n=6; Grade 2, n=4) and expected cytopenias. No patient died during the first 100 days after T-cell therapy. No neurotoxicity was observed. No objective responses were noted. Prolonged disease stabilization was noted in three patients lasting for 13.7, 12.9, and 7.3 months. High frequencies of target-specific T cells (up to 78.7% of CD8+ cells) were detected in the blood of treated patients, persisted for >1 year, and were detectable in post-treatment tumor tissue. Individual TCRs contained in T-cell products exhibited broad variation in TCR avidity, with the majority being of low-avidity. High-avidity TCRs were identified in some patients’ products. This study demonstrates the feasibility and tolerability of an actively personalized ACT directed to multiple defined pHLA cancer targets. Results warrant further evaluation of multi-target ACT approaches using potent high-avidity TCRs.
- Published
- 2023
7. Data from Phase I/II Multicenter Trial of a Novel Therapeutic Cancer Vaccine, HepaVac-101, for Hepatocellular Carcinoma
- Author
-
Luigi Buonaguro, Hans-Georg Rammensee, Harpreet Singh-Jasuja, Roberto S. Accolla, Toni Weinschenk, Carsten Reinhardt, Ulrike Gnad-Vogt, Mercedes Iñarrairaegui, Roberta Penta, Caterina Fusco, Maria Tagliamonte, Greta Forlani, Cécile Gouttefangeas, Regina Heidenreich, Tanguy Chaumette, Danila Valmori, Marco Borrelli, Heiko Schuster, Regina Mendrzyk, Katrin Aslan, Christian Flohr, Diego Duarte Alcoba, Jörg Ludwig, Antonio Avallone, Alessandro Inno, Luisa Vonghia, Sven Francque, Bruno Sangro, Yuk Ting Ma, Alfred Königsrainer, Paolo A. Ascierto, Andrea Mayer-Mokler, Francesco Izzo, Stefania Gori, and Markus W. Löffler
- Abstract
Purpose:Immunotherapy for hepatocellular carcinoma (HCC) shows considerable promise in improving clinical outcomes. HepaVac-101 represents a single-arm, first-in-human phase I/II multicenter cancer vaccine trial for HCC (NCT03203005). It combines multipeptide antigens (IMA970A) with the TLR7/8/RIG I agonist CV8102. IMA970A includes 5 HLA-A*24 and 7 HLA-A*02 as well as 4 HLA-DR restricted peptides selected after mass spectrometric identification in human HCC tissues or cell lines. CV8102 is an RNA-based immunostimulator inducing a balanced Th1/Th2 immune response.Patients and Methods:A total of 82 patients with very early- to intermediate-stage HCCs were enrolled and screened for suitable HLA haplotypes and 22 put on study treatment. This consisted in a single infusion of low-dose cyclophosphamide followed by nine intradermal coadministrations of IMA970A and CV8102. Only patients with no disease relapse after standard-of-care treatments were vaccinated. The primary endpoints of the HepaVac-101 clinical trial were safety, tolerability, and antigen-specific T-cell responses. Secondary or exploratory endpoints included additional immunologic parameters and survival endpoints.Results:The vaccination showed a good safety profile. Transient mild-to-moderate injection-site reactions were the most frequent IMA970A/CV8102-related side effects. Immune responses against ≥1 vaccinated HLA class I tumor-associated peptide (TAA) and ≥1 vaccinated HLA class II TAA were respectively induced in 37% and 53% of the vaccinees.Conclusions:Immunotherapy may provide a great improvement in treatment options for HCC. HepaVac-101 is a first-in-human clinical vaccine trial with multiple novel HLA class I– and class II–restricted TAAs against HCC. The results are initial evidence for the safety and immunogenicity of the vaccine. Further clinical evaluations are warranted.
- Published
- 2023
8. Supplementary Data from Phase I/II Multicenter Trial of a Novel Therapeutic Cancer Vaccine, HepaVac-101, for Hepatocellular Carcinoma
- Author
-
Luigi Buonaguro, Hans-Georg Rammensee, Harpreet Singh-Jasuja, Roberto S. Accolla, Toni Weinschenk, Carsten Reinhardt, Ulrike Gnad-Vogt, Mercedes Iñarrairaegui, Roberta Penta, Caterina Fusco, Maria Tagliamonte, Greta Forlani, Cécile Gouttefangeas, Regina Heidenreich, Tanguy Chaumette, Danila Valmori, Marco Borrelli, Heiko Schuster, Regina Mendrzyk, Katrin Aslan, Christian Flohr, Diego Duarte Alcoba, Jörg Ludwig, Antonio Avallone, Alessandro Inno, Luisa Vonghia, Sven Francque, Bruno Sangro, Yuk Ting Ma, Alfred Königsrainer, Paolo A. Ascierto, Andrea Mayer-Mokler, Francesco Izzo, Stefania Gori, and Markus W. Löffler
- Abstract
Supplementary Data from Phase I/II Multicenter Trial of a Novel Therapeutic Cancer Vaccine, HepaVac-101, for Hepatocellular Carcinoma
- Published
- 2023
9. Data from Targeting Carcinoembryonic Antigen with DNA Vaccination: On-Target Adverse Events Link with Immunologic and Clinical Outcomes
- Author
-
Christian H. Ottensmeier, Freda K. Stevenson, Gareth J. Thomas, Ugur Sahin, Petra Simon, Toni Weinschenk, Duncan I. Jodrell, Alan Anthoney, Sally Clive, Ann O'Callaghan, Andrew Bateman, Sarah Halford, Ceri Edwards, Emily Buxton, Paul Lloyd-Evans, Andrew King, Stephen M. Thirdborough, Jana Stasakova, Lindsey Chudley, Angelica Cazaly, Ann Mander, and Katy J. McCann
- Abstract
Purpose: We have clinically evaluated a DNA fusion vaccine to target the HLA-A*0201–binding peptide CAP-1 from carcinoembryonic antigen (CEA605–613) linked to an immunostimulatory domain (DOM) from fragment C of tetanus toxin.Experimental Design: Twenty-seven patients with CEA-expressing carcinomas were recruited: 15 patients with measurable disease (arm-I) and 12 patients without radiological evidence of disease (arm-II). Six intramuscular vaccinations of naked DNA (1 mg/dose) were administered up to week 12. Clinical and immunologic follow-up was up to week 64 or clinical/radiological disease.Results: DOM-specific immune responses demonstrated successful vaccine delivery. All patients without measurable disease compared with 60% with advanced disease responded immunologically, while 58% and 20% expanded anti-CAP-1 CD8+ T cells, respectively. CAP-1–specific T cells were only detectable in the blood postvaccination but could also be identified in previously resected cancer tissue. The gastrointestinal adverse event diarrhea was reported by 48% of patients and linked to more frequent decreases in CEA (P < 0.001) and improved global immunologic responses [anti-DOM responses of greater magnitude (P < 0.001), frequency (P = 0.004), and duration] compared with patients without diarrhea. In advanced disease patients, decreases in CEA were associated with better overall survival (HR = 0.14, P = 0.017). CAP-1 peptide was detectable on MHC class I of normal bowel mucosa and primary colorectal cancer tissue by mass spectrometry, offering a mechanistic explanation for diarrhea through CD8+ T-cell attack.Conclusions: Our data suggest that DNA vaccination is able to overcome peripheral tolerance in normal and tumor tissue and warrants testing in combination studies, for example, by vaccinating in parallel to treatment with an anti-PD1 antibody. Clin Cancer Res; 22(19); 4827–36. ©2016 AACR.
- Published
- 2023
10. Supplementary Figures 1-3 from Tumor-Associated MICA Is Shed by ADAM Proteases
- Author
-
Alexander Steinle, Hans-Georg Rammensee, Stefan Stevanovic, Andreas Ludwig, Mareike Wittenbrink, Toni Weinschenk, Friederike Gieseke, Dennis Goehlsdorf, and Inja Waldhauer
- Abstract
Supplementary Figures 1-3 from Tumor-Associated MICA Is Shed by ADAM Proteases
- Published
- 2023
11. Data from Tumor-Associated MICA Is Shed by ADAM Proteases
- Author
-
Alexander Steinle, Hans-Georg Rammensee, Stefan Stevanovic, Andreas Ludwig, Mareike Wittenbrink, Toni Weinschenk, Friederike Gieseke, Dennis Goehlsdorf, and Inja Waldhauer
- Abstract
The immunoreceptor NKG2D promotes immunosurveillance of malignant cells and protects the host from tumor initiation by activating natural killer cells and costimulating CD8 T cells. NKG2D-mediated recognition of malignant cells by cytotoxic lymphocytes is enabled through the tumor-associated expression of NKG2D ligands (NKG2DL) resulting from cellular or genotoxic stress. Shedding of NKG2DL is thought to constitute a major countermechanism of tumor cells to subvert NKG2D-mediated immunosurveillance. Here, we report that the prototypical NKG2DL MICA is released by proteolytic cleavage in the stalk of the MICA ectodomain, where deletions, but not alanine substitutions, impede MICA shedding. Small compound-mediated stimulation and inhibition of MICA shedding adduced characteristics that indicated an involvement of members of the “a disintegrin and metalloproteinase” (ADAM) family. Accordingly, MICA shedding by tumor cells was inhibited by silencing of the related ADAM10 and ADAM17 proteases, which are known to promote tumor growth by releasing epidermal growth factor receptor ligands. Collectively, our data show that ADAM10 and ADAM17 are critically involved in the tumor-associated proteolytic release of soluble MICA facilitating tumor immune escape. Hence, therapeutic blockade of ADAM10 and ADAM17 seems promising for cancer treatment by targeting both growth and immune escape of tumors. [Cancer Res 2008;68(15):6368–76]
- Published
- 2023
12. 1319 Next-generation TCR bispecifics (TCER®) targeting peptide-HLA antigens for the treatment of patients with solid tumors
- Author
-
Sebastian Bunk, Martin Hofmann, Gabriele Pszolla, Meike Hutt, Felix Unverdorben, Frank Schwoebel, Nadine Aschmoneit, Claudia Wagner, Maike Jaworski, Christoph Schraeder, Heiko Schuster, Sarah Missel, Toni Weinschenk, Dominik Maurer, and Carsten Reinhardt
- Published
- 2022
13. 713 The PRAME opportunity – high peptide copy numbers, homogenous expression and high prevalence to address a broad patient population across different solid cancers with TCR-based therapeutics
- Author
-
Cedrik Britten, Dejka Araujo, Linus Backert, Carsten Bokemeyer, Richard Carvajal, Manik Chatterjee, Asim Dash, Lena Freudenmann, Jens Fritsche, David Fuhrmann, Valentina Goldfinger, Tobias Holderried, Franziska Hoffgaard, Jens Hukelmann, Amir Jazaeri, Ahmed Kaseb, Florian Köhler, Daniel Kowalewski, Jason Luke, Van Morris, Shivani Mukhi, Martina Ott, Ran Reshef, Michael Römer, Lida Rostock, Swapna Satam, Arun Satelli, Christoph Schräder, Merline Thambi, Apostolia Tsimberidou, Maike Wagner, Martin Wermke, Heiko Schuster, Oliver Schoor, and Toni Weinschenk
- Published
- 2022
14. Quantitative immunopeptidomics reveals a tumor stroma-specific target for T cell therapy
- Author
-
Gloria B. Kim, Jens Fritsche, Sebastian Bunk, Andrea Mahr, Felix Unverdorben, Kevin Tosh, Hong Kong, Colby R. Maldini, Chui Lau, Sriram Srivatsa, Shuguang Jiang, Joshua Glover, Derek Dopkin, Carolyn X. Zhang, Heiko Schuster, Daniel J. Kowalewski, Valentina Goldfinger, Martina Ott, David Fuhrmann, Maike Baues, Hans Boesmueller, Christoph Schraeder, Gisela Schimmack, Colette Song, Franziska Hoffgaard, Michael Roemer, Chih-Chiang Tsou, Martin Hofmann, Thomas Treiber, Meike Hutt, Leonie Alten, Maike Jaworski, Amir Alpert, Sarah Missel, Carsten Reinhardt, Harpreet Singh, Oliver Schoor, Steffen Walter, Claudia Wagner, Dominik Maurer, Toni Weinschenk, and James L. Riley
- Subjects
Proteomics ,Antigens, Neoplasm ,Cell Line, Tumor ,T-Lymphocytes ,Cell- and Tissue-Based Therapy ,Receptors, Antigen, T-Cell ,Humans ,General Medicine ,Immunotherapy, Adoptive ,Article - Abstract
T cell receptor (TCR)–based immunotherapy has emerged as a promising therapeutic approach for the treatment of patients with solid cancers. Identifying peptide–human leukocyte antigen (pHLA) complexes highly presented on tumors and rarely expressed on healthy tissue in combination with high-affinity TCRs that when introduced into T cells can redirect T cells to eliminate tumor but not healthy tissue is a key requirement for safe and efficacious TCR-based therapies. To discover promising shared tumor antigens that could be targeted via TCR-based adoptive T cell therapy, we employed population-scale immunopeptidomics using quantitative mass spectrometry across ~1500 tumor and normal tissue samples. We identified an HLA-A*02:01-restricted pan-cancer epitope within the collagen type VI α-3 ( COL6A3 ) gene that is highly presented on tumor stroma across multiple solid cancers due to a tumor-specific alternative splicing event that rarely occurs outside the tumor microenvironment. T cells expressing natural COL6A3-specific TCRs demonstrated only modest activity against cells presenting high copy numbers of COL6A3 pHLAs. One of these TCRs was affinity-enhanced, enabling transduced T cells to specifically eliminate tumors in vivo that expressed similar copy numbers of pHLAs as primary tumor specimens. The enhanced TCR variants exhibited a favorable safety profile with no detectable off-target reactivity, paving the way to initiate clinical trials using COL6A3-specific TCRs to target an array of solid tumors.
- Published
- 2022
15. IMA402, an Off-the-Shelf, Next-Generation TCR Bispecific (TCER®) for Efficiently Targeting an HLA-Presented Peptide from the Pan-Cancer Antigen PRAME
- Author
-
Sebastian Bunk, Martin Hofmann, Gabriele Pszolla, Meike Hutt, Frank Schwöbel, Felix Unverdorben, Nadine Aschmoneit, Claudia Wagner, Maike Jaworski, Heiko Schuster, Florian Schwoerer, Christoph Schraeder, Oliver Schoor, Sarah Missel, Toni Weinschenk, Dominik Maurer, and Carsten Reinhardt
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
16. Phase I/II multicenter trial of a novel therapeutic cancer vaccine, HepaVac-101, for hepatocellular carcinoma
- Author
-
Markus W. Löffler, Stefania Gori, Francesco Izzo, Andrea Mayer-Mokler, Paolo A. Ascierto, Alfred Königsrainer, Yuk Ting Ma, Bruno Sangro, Sven Francque, Luisa Vonghia, Alessandro Inno, Antonio Avallone, Jörg Ludwig, Diego Duarte Alcoba, Christian Flohr, Katrin Aslan, Regina Mendrzyk, Heiko Schuster, Marco Borrelli, Danila Valmori, Tanguy Chaumette, Regina Heidenreich, Cécile Gouttefangeas, Greta Forlani, Maria Tagliamonte, Caterina Fusco, Roberta Penta, Mercedes Iñarrairaegui, Ulrike Gnad-Vogt, Carsten Reinhardt, Toni Weinschenk, Roberto S. Accolla, Harpreet Singh-Jasuja, Hans-Georg Rammensee, and Luigi Buonaguro
- Subjects
Cancer Research ,Tumor-associated antigens ,Carcinoma, Hepatocellular ,cancer immunotherapy ,HLA-A Antigens ,Hepatocellular carcinoma ,Liver Neoplasms ,Cancer Vaccines ,Adjuvants, Immunologic ,Oncology ,Humans ,Immunotherapy ,Human medicine ,Peptides ,cancer vaccine - Abstract
Purpose: Immunotherapy for hepatocellular carcinoma (HCC) shows considerable promise in improving clinical outcomes. HepaVac-101 represents a single-arm, first-in-human phase I/II multicenter cancer vaccine trial for HCC (NCT03203005). It combines multipeptide antigens (IMA970A) with the TLR7/8/RIG I agonist CV8102. IMA970A includes 5 HLA-A*24 and 7 HLA-A*02 as well as 4 HLA-DR restricted peptides selected after mass spectrometric identification in human HCC tissues or cell lines. CV8102 is an RNA-based immunostimulator inducing a balanced Th1/Th2 immune response. Patients and Methods: A total of 82 patients with very early- to intermediate-stage HCCs were enrolled and screened for suitable HLA haplotypes and 22 put on study treatment. This consisted in a single infusion of low-dose cyclophosphamide followed by nine intradermal coadministrations of IMA970A and CV8102. Only patients with no disease relapse after standard-of-care treatments were vaccinated. The primary endpoints of the HepaVac-101 clinical trial were safety, tolerability, and antigen-specific T-cell responses. Secondary or exploratory endpoints included additional immunologic parameters and survival endpoints. Results: The vaccination showed a good safety profile. Transient mild-to-moderate injection-site reactions were the most frequent IMA970A/CV8102-related side effects. Immune responses against ≥1 vaccinated HLA class I tumor-associated peptide (TAA) and ≥1 vaccinated HLA class II TAA were respectively induced in 37% and 53% of the vaccinees. Conclusions: Immunotherapy may provide a great improvement in treatment options for HCC. HepaVac-101 is a first-in-human clinical vaccine trial with multiple novel HLA class I– and class II–restricted TAAs against HCC. The results are initial evidence for the safety and immunogenicity of the vaccine. Further clinical evaluations are warranted.
- Published
- 2022
17. Immunopeptidomics and Peptide Expression Profiles to Develop T-Cell Receptors Against Glioma-Associated Antigens
- Author
-
Payal Watchmaker, Diego Carrera, Joseph F. Costello, Michael C. Oldham, Harpreet Singh, Samuel J. Shelton, Oliver Schoor, Josie Hayes, Nolbert Hilf, Gary Kohanbash, Hideho Okada, Sören Müller, and Toni Weinschenk
- Subjects
business.industry ,medicine.medical_treatment ,Alternative splicing ,T-cell receptor ,Human leukocyte antigen ,Immunotherapy ,medicine.disease ,Epitope ,Antigen ,Glioma ,medicine ,Cancer research ,Surgery ,Neurology (clinical) ,business ,Gene - Published
- 2019
18. 293 Resultsof the first-in-human clinical trial with personalized multi-target adoptive cell therapy (ACTolog IMA101)
- Author
-
Dominik Maurer, Borje S. Andersson, Jens Fritsche, Arun Satelli, Sabrina Kuttruff-Coqui, Kerstin Guenther, Carsten Reinhardt, Steffen Walter, Mamta Kalra, Ali Mohamed, Apostolia Maria Tsimberidou, Rita Ort, Kerry Sieger, Norbert Hilf, Amir Alpert, Anna Nowak, Becky Norris, Toni Weinschenk, David Vining, Oliver Schoor, Fabian Richter, Patrick Hwu, Zoe Coughlin, Cassian Yee, Regina Mendrzyk, Harpreet Singh, and Claudia Wagner
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Context (language use) ,Leukapheresis ,medicine.disease ,Clinical trial ,Cytokine release syndrome ,Regimen ,Tolerability ,Atezolizumab ,Internal medicine ,medicine ,Clinical endpoint ,business - Abstract
Background ACTolog (IMA101) is a personalized multi-target adoptive cell therapy (ACT) approach in which autologous T-cell products are redirected against multiple novel defined peptide-HLA (pHLA) cancer targets identified by the target discovery platform XPRESIDENT®. The primary endpoint was to assess the safety of ACTolog. Secondary endpoints were to assess the in vivo persistence of transferred T-cells and antitumor activity (www.clinicaltrials.gov NCT02876510). Methods HLA-A*02:01 positive patients with relapsed/refractory solid tumors whose tumor expressed ≥1 cancer target underwent leukapheresis and endogenous T-cells specific for up to 4 targets were primed and expanded in vitro. Patients received lymphodepletion (fludarabine 40 mg/m² and cyclophosphamide 500 mg/m² on days -6 to -3) followed by up to 4 × 1010 cells (day 0), and IL-2 (1 × 106 IU/m² SC twice daily, 14 days) (Cohort 1). In addition, cohort 2 received atezolizumab (1200 mg IV) every 21 days upon hematologic recovery. Infused T-cells were tracked in patients‘ blood via flow cytometry-based immunomonitoring as well as TCRβ sequencing. TCRs from target specific T-cells were identified from patients‘ T-cell products and characterized. Results From 07/2017–03/2020, 214 patients were screened, and 14 heavily pre-treated patients with various tumor types were infused with 1–3 T-cell products each (table 1). The treatment was generally well tolerated. The most common adverse events observed to date were expected cytopenias, mostly attributed to the lymphodepleting regimen, and Grade 1–2 cytokine release syndrome. Prolonged disease stabilization was noted in three patients for 12 months, 12+ months, and 7 months. High frequencies of target-specific T-cells up to 78.7% of CD8+ cells were detected in the blood of treated patients, persisted for >1 year and were detected in post-treatment tumor biopsies. Characterization of individual TCRs contained in T-cell products showed a broad variation of TCR avidities with the majority of TCRs being of low avidity. High-avidity TCRs were identified from products of some patients, including a patient with 26% decrease in tumor measurements 6 weeks post-treatment. Tracking the respective T-cell clonotypes in patients‘ blood and tumor provides insights into the mechanism of tumor control. Six-month data will be presented at the conference. Conclusions This is the first reported trial demonstrating the feasibility and tolerability of a personalized ACT approach using multiple defined T-cell products directed to multiple precisely defined pHLA cancer targets. These results support further exploration of a multi-target ACT approach, particularly in the context of T-cells expressing high-avidity TCRs to such defined pHLA targets. Trial Registration https://clinicaltrials.gov/ct2/show/NCT02876510 Ethics Approval The study was approved by WCG WIRB, IRB tracking number 20162235. The study was conducted in accordance with the Declaration of Helsinki and the International Conference on Harmonization Good Clinical Practice guidelines. All the study participants provided written informed consent before enrollment. Consent Patient consent for publication is not required. Patients consented to participate in the study.
- Published
- 2020
19. 959 Safety and anti-tumor activity of TCR-engineered autologous, PRAME-directed T cells across multiple advanced solid cancers at low doses – clinical update on the ACTengine® IMA203 trial
- Author
-
Cedrik M. Britten, Regina Mendrzyk, Manik Chatterjee, Tobias A. W. Holderried, Norbert Hilf, Van K. Morris, Andrea Mayer-Mokler, Dominik Maurer, Jason J. Luke, Martin Wermke, Kerstin Guenther, Carsten Reinhardt, Stephen Eck, Dejka M. Araujo, Mamta Kalra, Harpreet Singh, Toni Weinschenk, George Jr Blumenschein, Ali Mohamed, Arun Satelli, Apostolia-Maria Tsimberidou, Winfried Alsdorf, and Steffen Walter
- Subjects
Pharmacology ,Oncology ,Cancer Research ,medicine.medical_specialty ,PRAME ,Cyclophosphamide ,business.industry ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Leukapheresis ,Fludarabine ,Cell therapy ,Tolerability ,Internal medicine ,medicine ,Molecular Medicine ,Immunology and Allergy ,Cytotoxic T cell ,Cancer/testis antigens ,business ,RC254-282 ,medicine.drug - Abstract
BackgroundAdoptive cell therapy demonstrated significant clinical benefit in patients with hematological malignancies but results in most solid tumors have been less encouraging so far.In the IMA203 trial we are treating advanced solid cancer patients utilizing TCR-engineered T cells (TCR-T) directed against an HLA-A*02-restricted peptide derived from the highly prevalent cancer testis antigen PRAME. This target was selected due to homogenous expression and exceptionally high target peptide density per tumor cell (assessed by quantitative mass spectrometry), two features we hypothesize to be critical determinants of anti-tumor activity in TCR-T trials.MethodsThis ongoing first-in-human, dose escalation, multi-indication trial enrolls HLA-A*02:01- and PRAME-positive recurrent and/or refractory solid cancer patients, who failed all available standard treatments. Eligible patients undergo leukapheresis and an autologous TCR-T product is manufactured. After lymphodepletion with fludarabine and cyclophosphamide, T cells are infused, followed by low-dose IL-2. The primary objective of the trial is to assess the safety and tolerability of IMA203. Secondary objectives are to evaluate the anti-tumor activity and pharmacodynamics using molecular and immunological methods.ResultsAs of August 15, 2021, 16 heavily pre-treated patients received IMA203 T cells across multiple escalating dose levels (DL). Absolute IMA203 doses infused ranged from 0.08 to 0.81x109 transduced CD8 T cells per patient, which to our knowledge did not lead to anti-tumor responses in other TCR-T trials. Treatment-emergent adverse events after IMA203 infusion were transient and manageable. Most common events were expected cytopenias (G1-4), CRS and ICANS (both G1-2) and 1 DLT in DL2 (reported earlier). All evaluable patients (N=12) achieved disease control (i.e. best overall response: stable disease [SD] or partial response [PR]) and 6 patients demonstrated PRs according to RECIST1.1 with 2 of these PRs being confirmed. While all 3 patients treated at DL1 (median dose: 0.11x109) experienced SD, a PR was observed in 6/9 patients treated beyond DL1 (median dose: 0.30x109). Responses were seen in patients with synovial sarcoma (N=3), malignant melanoma (N=2) and head and neck cancer (N=1). Robust engraftment of T cells was observed in all patients and tumor infiltration by TCR-modified T cells was demonstrated in patients with evaluable on-treatment biopsies.ConclusionsTo our knowledge IMA203 is the first TCR-T product candidate that induced frequent tumor responses across multiple solid cancers using transduced T cells at doses below 1 billion and has a manageable safety profile. The next step is to assess response rates at higher dose levels and durability of responses.Trial RegistrationNCT03686124Ethics ApprovalThe study was approved by the institutional review board/ethics committee as required for each participating site.
- Published
- 2021
20. Immunogenic stress and death of cancer cells: Contribution of antigenicity vs adjuvanticity to immunosurveillance
- Author
-
Antonella Sistigu, Steffen Walter, Juliette Humeau, Aitziber Buqué, Jan W. Drijfhout, Laura Senovilla, Norma Bloy, Claude Perreault, Guido Kroemer, Pauline Garcia, Jonathan Pol, Céline M. Laumont, Eric Bonneil, Jonathan M. Pitt, Takahiro Yamazaki, Laurence Zitvogel, Hans-Georg Rammensee, Jens Fritsche, Toni Weinschenk, Pierre Thibault, Cornelis J. M. Melief, Gautier Stoll, and Guillaume Meurice
- Subjects
0301 basic medicine ,autophagy ,Programmed cell death ,immunopeptidome ,Immunology ,Antigen presentation ,hyperploidy ,Major histocompatibility complex ,calreticulin ,Mice ,03 medical and health sciences ,Adenosine Triphosphate ,Immune system ,Adjuvants, Immunologic ,Antigen ,Antigens, Neoplasm ,Monitoring, Immunologic ,Neoplasms ,Animals ,Humans ,Immunology and Allergy ,Immunologic Surveillance ,Cell Death ,biology ,Autophagy ,food and beverages ,Cell biology ,Immunosurveillance ,030104 developmental biology ,Cancer cell ,endoplasmic reticulum stress ,biology.protein ,Signal Transduction - Abstract
Cancer cells are subjected to constant selection by the immune system, meaning that tumors that become clinically manifest have managed to subvert or hide from immunosurveillance. Immune control can be facilitated by induction of autophagy, as well as by polyploidization of cancer cells. While autophagy causes the release of ATP, a chemotactic signal for myeloid cells, polyploidization can trigger endoplasmic reticulum stress with consequent exposure of the "eat-me" signal calreticulin on the cell surface, thereby facilitating the transfer of tumor antigens into dendritic cells. Hence, both autophagy and polyploidization cause the emission of adjuvant signals that ultimately elicit immune control by CD8+ T lymphocytes. We investigated the possibility that autophagy and polyploidization might also affect the antigenicity of cancer cells by altering the immunopeptidome. Mass spectrometry led to the identification of peptides that were presented on major histocompatibility complex (MHC) class I molecules in an autophagy-dependent fashion or that were specifically exposed on the surface of polyploid cells, yet lost upon passage of such cells through immunocompetent (but not immunodeficient) mice. However, the preferential recognition of autophagy-competent and polyploid cells by the innate and cellular immune systems did not correlate with the preferential recognition of such peptides in vivo. Moreover, vaccination with such peptides was unable to elicit tumor growth-inhibitory responses in vivo. We conclude that autophagy and polyploidy increase the immunogenicity of cancer cells mostly by affecting their adjuvanticity rather than their antigenicity.
- Published
- 2017
21. Actively personalized vaccination trial for newly diagnosed glioblastoma
- Author
-
Marij J. P. Welters, Dominik Maurer, Ulrik Lassen, Martin Löwer, Bernhard Rossler, Ugur Sahin, Andreas von Deimling, Toni Weinschenk, Christian H. Ottensmeier, Elisa Rusch, Colette Song, Valérie Dutoit, Jordi Rodon, Norbert Hilf, Hans Skovgaard Poulsen, Nina Pawlowski, Francisco Martínez-Ricarte, Judith R. Kroep, Juan Sahuquillo, Claudia Wagner, Edward W. Green, Sonja Dorner, Cedrik M. Britten, Franziska Hoffgaard, Jens Fritsche, Ghazaleh Tabatabai, Stefan Stevanovic, Harpreet Singh-Jasuja, Marco Skardelly, Sabrina Kuttruff-Coqui, Hans-Georg Rammensee, Katharina Kiesel, Alexander Ulges, Carsten Reinhardt, Michael Platten, Alexandra Kemmer-Brück, Bracha Shraibman, Denis Migliorini, Sebastian Kreiter, Jordi Piro, Oliver Schoor, Valesca Bukur, Katrin Frenzel, Berta Ponsati, David Capper, Jorg Ludwig, Monika Stieglbauer, Regina Mendrzyk, Miriam Meyer, Sjoerd H. van der Burg, Evelyna Derhovanessian, Pierre-Yves Dietrich, Arie Admon, Arbel D. Tadmor, Manja Idorn, Wolfgang Wick, Hideho Okada, Per thor Straten, Sandra Heesch, Lukas Bunse, Christoph Huber, Katy J. McCann, Cécile Gouttefangeas, John C. Castle, Dutoit Vallotton, Valérie, Migliorini, Denis, and Dietrich, Pierre-Yves
- Subjects
Adult ,Male ,0301 basic medicine ,T-Lymphocytes Helper-Inducer/immunology ,T cell ,Antigens Neoplasm/immunology ,Epitopes, T-Lymphocyte ,Human leukocyte antigen ,CD8-Positive T-Lymphocytes ,CD8-Positive T-Lymphocytes/immunology ,Cancer Vaccines ,Epitopes ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Glioblastoma/diagnosis/immunology/therapy ,Antigen ,Antigens, Neoplasm ,Glioma ,medicine ,Humans ,Precision Medicine ,Aged ,ddc:616 ,Cancer Vaccines/immunology/therapeutic use ,HLA-A Antigens/immunology ,Multidisciplinary ,HLA-A Antigens ,business.industry ,T-Lymphocyte/immunology ,Immunogenicity ,T-Lymphocytes, Helper-Inducer ,Precision Medicine/methods ,Middle Aged ,medicine.disease ,Vaccination ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Immunologic Memory/immunology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Glioblastoma ,business ,Immunologic Memory ,CD8 - Abstract
Patients with glioblastoma currently do not sufficiently benefit from recent breakthroughs in cancer treatment that use checkpoint inhibitors1,2. For treatments using checkpoint inhibitors to be successful, a high mutational load and responses to neoepitopes are thought to be essential3. There is limited intratumoural infiltration of immune cells4 in glioblastoma and these tumours contain only 30–50 non-synonymous mutations5. Exploitation of the full repertoire of tumour antigens—that is, both unmutated antigens and neoepitopes—may offer more effective immunotherapies, especially for tumours with a low mutational load. Here, in the phase I trial GAPVAC-101 of the Glioma Actively Personalized Vaccine Consortium (GAPVAC), we integrated highly individualized vaccinations with both types of tumour antigens into standard care to optimally exploit the limited target space for patients with newly diagnosed glioblastoma. Fifteen patients with glioblastomas positive for human leukocyte antigen (HLA)-A*02:01 or HLA-A*24:02 were treated with a vaccine (APVAC1) derived from a premanufactured library of unmutated antigens followed by treatment with APVAC2, which preferentially targeted neoepitopes. Personalization was based on mutations and analyses of the transcriptomes and immunopeptidomes of the individual tumours. The GAPVAC approach was feasible and vaccines that had poly-ICLC (polyriboinosinic-polyribocytidylic acid-poly-l-lysine carboxymethylcellulose) and granulocyte–macrophage colony-stimulating factor as adjuvants displayed favourable safety and strong immunogenicity. Unmutated APVAC1 antigens elicited sustained responses of central memory CD8+ T cells. APVAC2 induced predominantly CD4+ T cell responses of T helper 1 type against predicted neoepitopes.
- Published
- 2019
22. Pitfalls in HLA Ligandomics—How to Catch a Li(e)gand
- Author
-
Linus Backert, Martin Priemer, Oliver Schoor, Michael Römer, Jens Fritsche, Toni Weinschenk, Chih-Chiang Tsou, Heiko Schuster, Franziska Hoffgaard, Sonja Dorner, Frederik Gwinner, and Daniel J. Kowalewski
- Subjects
Proteomics ,False discovery rate ,FDR, false discovery rate ,Proteome ,CID, collision-induced dissociation ,Computer science ,medicine.medical_treatment ,PSM, peptide–spectrum match ,Peptide ,Computational biology ,Human leukocyte antigen ,Ligands ,Biochemistry ,Analytical Chemistry ,SIL, stable isotope label ,SST, system suitability testing ,03 medical and health sciences ,Special Issue: Immunopeptidomics ,Cancer immunotherapy ,HLA Antigens ,PRM, parallel reaction monitoring ,peptide quality control ,medicine ,Humans ,IS-PRM, internal standard triggered PRM ,SFF, source fragmentation fraction ,Molecular Biology ,PSSM, position-specific scoring matrix ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,cancer immunotherapy ,HLA, human leukocyte antigen ,Repertoire ,TCR targets ,030302 biochemistry & molecular biology ,Technological Innovation and Resources ,immunopeptidomics ,gRT, globally aligned retention time ,HCD, higher collisional energy dissociation ,Identification (information) ,PCC, proteolytic contamination count ,chemistry ,Proteolysis ,HLA ligands ,Experimental methods ,Peptides - Abstract
Knowledge about the peptide repertoire presented by human leukocyte antigens (HLA) holds the key to unlock target-specific cancer immunotherapies such as adoptive cell therapies or bispecific T cell engaging receptors. Therefore, comprehensive and accurate characterization of HLA peptidomes by mass spectrometry (immunopeptidomics) across tissues and disease states is essential. With growing numbers of immunopeptidomics datasets and the scope of peptide identification strategies reaching beyond the canonical proteome, the likelihood for erroneous peptide identification as well as false annotation of HLA-independent peptides as HLA ligands is increasing. Such “fake ligands” can lead to selection of nonexistent targets for immunotherapeutic development and need to be recognized as such as early as possible in the preclinical pipeline. Here we present computational and experimental methods that enable the identification of “fake ligands” that might be introduced at different steps of the immunopeptidomics workflow. The statistics presented herein allow discrimination of true HLA ligands from coisolated HLA-independent proteolytic fragments. In addition, we describe necessary steps to ensure system suitability of the chromatographic system. Furthermore, we illustrate an algorithm for detection of source fragmentation events that are introduced by electrospray ionization during mass spectrometry. For confirmation of peptide sequences, we present an experimental pipeline that enables high-throughput sequence verification through similarity of fragmentation pattern and coelution of synthetic isotope-labeled internal standards. Based on these methods, we show the overall high quality of existing datasets but point out limitations and pitfalls critical for individual peptides and how they can be uncovered in order to identify true ligands., Graphical Abstract, Highlights • Best practices to identify true HLA ligands as targets for cancer immunotherapies. • Quality control in mass spectrometry to improve neoantigen/crypto-target discovery. • Computational methods to assess fragment contamination in immunopeptidomics. • Experimental methods for LC system suitability testing and HT sequence verification., In Brief Accurate characterization of the HLA ligandome by mass spectrometry holds the key to unlock target-specific cancer immunotherapies such as adoptive cell therapies or bispecific T cell engaging receptors. Quality control at each step of the immunopeptidomics pipeline is relevant to differentiate between false and true HLA ligands. Here, we present computational and experimental methods as part of our XPRESIDENT technology platform that allow identification of true HLA ligands—an essential prerequisite for successful therapeutic development.
- Published
- 2021
23. IMA901, a multipeptide cancer vaccine, plus sunitinib versus sunitinib alone, as first-line therapy for advanced or metastatic renal cell carcinoma (IMPRINT): a multicentre, open-label, randomised, controlled, phase 3 trial
- Author
-
Tim Eisen, Mikhail Kogan, Arnulf Stenzl, Jens Bedke, Dominik Maurer, Simon J. Crabb, Mikhail Shkolnik, Alexandra Kirner, Stéphane Oudard, Steffen Walter, Romauld Zdrojowy, Carsten Reinhardt, Harpreet Singh-Jasuja, Jens Fritsche, Andrea Mahr, Steffen Weikert, Sergio Bracarda, Toni Weinschenk, Brian I. Rini, Regina Mendrzyk, Claudia Wagner, and Joerg Ludwig
- Subjects
Male ,medicine.medical_specialty ,Indoles ,medicine.medical_treatment ,Phases of clinical research ,Neutropenia ,urologic and male genital diseases ,Cancer Vaccines ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Renal cell carcinoma ,Internal medicine ,Sunitinib ,Clinical endpoint ,Carcinoma ,Humans ,Medicine ,Pyrroles ,Neoplasm Metastasis ,Carcinoma, Renal Cell ,Aged ,Intention-to-treat analysis ,business.industry ,Middle Aged ,medicine.disease ,Kidney Neoplasms ,Nephrectomy ,Surgery ,Oncology ,030220 oncology & carcinogenesis ,Female ,business ,030215 immunology ,medicine.drug - Abstract
Summary Background In a phase 2 study in patients with metastatic renal cell carcinoma, overall survival was associated with T-cell responses against IMA901, a vaccine consisting of ten tumour-associated peptides. In this phase 3 trial, we aimed to determine the clinical effect of adding IMA901 to sunitinib, the standard first-line treatment in metastatic renal cell carcinoma with postulated favourable immunomodulatory effects. Methods The IMPRINT study is an open-label, randomised, controlled, phase 3 trial done at 124 clinical sites in 11 countries. HLA-A*02 -positive patients (aged ≥18 years) with treatment-naive, histologically confirmed metastatic or locally advanced (or both) clear-cell renal cell carcinoma were randomly assigned (3:2) to receive sunitinib plus up to ten intradermal vaccinations of IMA901 (4·13 mg) and granulocyte macrophage colony-stimulating factor (75 μg), with one dose of cyclophosphamide (300 mg/m 2 ) 3 days before the first vaccination, or to receive sunitinib alone. Sunitinib (50 mg) was given orally once daily, with each cycle defined as 4 weeks on treatment followed by 2 weeks off treatment, until progression of disease as determined by the investigator, death, or withdrawal of consent. Block randomisation (block size five) was done centrally using an interactive web response system, stratified by prognostic risk, geographical region, and previous nephrectomy. Patients and investigators were not masked to treatment allocation. The primary endpoint was overall survival from randomisation until death of any cause as determined by the investigator, analysed by intention to treat. This study is registered with ClinicalTrials.gov, number NCT01265901. Findings Between Dec 22, 2010, and Dec 15, 2012, we screened 1171 patients, of whom 339 were randomly assigned to receive sunitinib plus IMA901 (n=204) or sunitinib monotherapy (n=135). Patients had a median follow-up of 33·27 months (IQR 29·92–35·64). Median overall survival did not differ significantly between the groups (33·17 months [95% CI 27·81–41·36] in the sunitinib plus IMA901 group vs not reached [33·67–not reached] in the sunitinib monotherapy group; hazard ratio 1·34 [0·96–1·86]; p=0·087). 116 (57%) of 202 patients in the sunitinib plus IMA901 group and 62 (47%) of 132 in the sunitinib group had grade 3 or worse adverse events, the most common of which were hypertension, neutropenia, and anaemia in both groups, and mild-to-moderate transient injection-site reactions (eg, erythema, pruritus) were the most frequent IMA901-related side-effect in the sunitinib plus IMA901 group. Serious adverse events leading to death occurred in four (2%) patients (one respiratory failure and circulatory collapse [possibly related to sunitinib], one oesophageal varices haemorrhage [possibly related to sunitinib], one cardiac arrest [possibly related to sunitinib], and one myocardial infarction) and eight (6%) patients in the sunitinib group (one case each of renal failure, oesophageal varices haemorrhage, circulatory collapse, wound infection, ileus, cerebrovascular accident [possibly treatment related], and sepsis). Interpretation IMA901 did not improve overall survival when added to sunitinib as first-line treatment in patients with metastatic renal cell carcinoma. The magnitude of immune responses needs to be improved before further development of IMA901 in this disease is indicated. Funding Immatics Biotechnologies.
- Published
- 2016
24. RNA editing derived epitopes function as cancer antigens to elicit immune responses
- Author
-
Patrick Hwu, Oliver Schoor, Yulun Chiu, Franziska Hoffgaard, Amjad H. Talukder, Xinxin Peng, Leng Han, Cara Haymaker, Han Liang, Kenneth L. Scott, Chantale Bernatchez, Jason Roszik, Kathleen Kong, Valentina Goldfinger, Leila Williams, Minying Zhang, Jens Fritsche, Greg Lizee, Marie Andrée Forget, Toni Weinschenk, Chih-Chiang Tsou, Harpreet Singh-Jasuja, Gordon B. Mills, Yiu Huen Tsang, and Xiaoyan Xu
- Subjects
Cytotoxicity, Immunologic ,0301 basic medicine ,Science ,General Physics and Astronomy ,Human leukocyte antigen ,CD8-Positive T-Lymphocytes ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Epitope ,Epitopes ,03 medical and health sciences ,Cyclin I ,0302 clinical medicine ,Immune system ,Antigen ,Antigens, Neoplasm ,HLA Antigens ,Cell Line, Tumor ,Neoplasms ,Humans ,lcsh:Science ,Cells, Cultured ,Proteogenomics ,Antigen Presentation ,Messenger RNA ,Multidisciplinary ,Effector ,RNA ,General Chemistry ,Cell biology ,030104 developmental biology ,RNA editing ,Immune System ,030220 oncology & carcinogenesis ,lcsh:Q ,RNA Editing ,Peptides - Abstract
In addition to genomic mutations, RNA editing is another major mechanism creating sequence variations in proteins by introducing nucleotide changes in mRNA sequences. Deregulated RNA editing contributes to different types of human diseases, including cancers. Here we report that peptides generated as a consequence of RNA editing are indeed naturally presented by human leukocyte antigen (HLA) molecules. We provide evidence that effector CD8+ T cells specific for edited peptides derived from cyclin I are present in human tumours and attack tumour cells that are presenting these epitopes. We show that subpopulations of cancer patients have increased peptide levels and that levels of edited RNA correlate with peptide copy numbers. These findings demonstrate that RNA editing extends the classes of HLA presented self-antigens and that these antigens can be recognised by the immune system.
- Published
- 2018
25. Effective Targeting of PRAME-Positive Tumors with Bispecific T Cell-Engaging Receptor (TCER®) Molecules
- Author
-
Oliver Schoor, Felix Unverdorben, Harpreet Singh-Jasuja, Heiko Schuster, Meike Hutt, Toni Weinschenk, Sebastian Bunk, Gabriele Pszolla, Claudia Wagner, Martin Hofmann, Carsten Reinhardt, Dominik Maurer, Frank Schwöbel, Sarah Missel, and Sara Yousef
- Subjects
PRAME ,biology ,Chemistry ,T cell ,Immunology ,T-cell receptor ,Cell Biology ,Hematology ,Human leukocyte antigen ,Biochemistry ,Fusion protein ,CD19 ,medicine.anatomical_structure ,Antigen ,Cancer cell ,medicine ,Cancer research ,biology.protein - Abstract
T cell receptors (TCRs) naturally recognize human leukocyte antigen (HLA)-bound peptides derived from foreign and endogenous proteins regardless of their extracellular or intracellular location. Preferentially expressed antigen in melanoma (PRAME) has been shown to be expressed at high levels in a variety of cancer cells while being absent or present only at very low levels in normal adult tissues except testis. In contrast to most other cancer/testis antigens, PRAME is expressed not only in solid tumors but also in leukemia and myeloma cells. Immunotherapy with bispecific T cell engagers has emerged as a novel and promising treatment modality for malignant diseases, however, antibody-based approaches (ie. blinatumomab) are restricted to few surface antigens such as CD19 or BCMA. Immatics has developed bispecific T cell-engaging receptors (TCER®) that are fusion proteins consisting of an affinity-maturated TCR and a humanized T cell-recruiting antibody with an effector function-silenced IgG1 Fc part. TCER® molecules confer extended half-life together with antibody-like stability and manufacturability characteristics. The molecular design allows for effective redirection of T cells towards target peptide-HLA selectively expressed in tumor tissues. Here we present proof-of-concept data from a TCER® program targeting a PRAME-derived peptide bound to HLA-A*02:01. We confirmed the abundant presence of the target peptide-HLA in several cancer indications and its absence in relevant human normal tissues by using the XPRESIDENT® target discovery engine, which combines quantitative mass spectrometry, transcriptomics and bioinformatics. Yeast surface display technology was used to maturate the stability and affinity of a parental human TCR recognizing PRAME with high functional avidity and specificity. During maturation we applied XPRESIDENT®-guided off-target toxicity screening, incorporating the world's largest normal tissue immunopeptidome database, to deselect cross-reactive candidate TCRs. The maturated TCRs were engineered into the TCER® scaffold and production in Chinese hamster ovary (CHO) cells generated highly stable molecules with low tendency for aggregation as confirmed during stress studies. Following TCR maturation, the TCER® molecules exhibited an up to 10,000-fold increased binding affinity towards PRAME when compared to the parental TCR. The high affinity correlated with potent in vitro anti-tumor activity requiring only low picomolar concentrations of TCER® molecules to induce half-maximal lysis of tumor cells expressing the target at physiological levels. Furthermore, using a tumor xenograft model in immunodeficient NOG mice, we could demonstrate significant growth inhibition of established tumors upon intravenous injection of TCER® molecules. Pharmacokinetic profiling in NOG mice determined a terminal half-life of more than 4 days, compatible with a once weekly dosing regimen in patients. For the safety assessment, we measured killing of more than 20 different human normal tissue cell types derived from high risk organs. Notably, we could confirm a favorable safety window for selected TCER® molecules, which induced killing of most normal tissue cells only at significantly higher concentrations than required for killing of tumor cells. To further support safety of TCER® molecules, we also performed a comprehensive characterization of potential off-target peptides selected from the XPRESIDENT® normal tissue database based on its high similarity to the sequence of the target peptide or based on data from alternative screening approaches. In summary, the efficacy, safety and manufacturability data to be presented provide preclinical proof-of-concept for a novel bispecific T cell-engaging receptor (TCER®) molecule targeting PRAME for treatment of various malignant diseases. Disclosures Bunk: Immatics: Employment. Hofmann:Immatics: Employment. Unverdorben:Immatics: Employment. Hutt:Immatics: Employment. Pszolla:Immatics: Employment. Schwöbel:Immatics: Employment. Wagner:Immatics: Employment. Yousef:Immatics: Employment. Schuster:Immatics: Employment. Missel:Immatics: Employment. Schoor:Immatics: Employment. Weinschenk:Immatics: Employment, Equity Ownership. Singh-Jasuja:Immatics: Employment, Equity Ownership. Maurer:Immatics: Employment. Reinhardt:Immatics: Employment, Equity Ownership.
- Published
- 2019
26. Abstract 662: IMA_Detect: Mass spectrometry guided development and clinical application of a companion diagnostic for adoptive cellular therapy against tumor associated HLA peptides
- Author
-
Fritsche, Jens, primary, Satelli, Arun, additional, Hörzer, Helen, additional, Rakitsch, Barbara, additional, Hoffgaard, Franziska, additional, Hilf, Norbert, additional, Schoor, Oliver, additional, Singh-Jasuja, Harpreet, additional, and Toni, Weinschenk, additional
- Published
- 2018
- Full Text
- View/download PDF
27. Targeting Carcinoembryonic Antigen with DNA Vaccination: On-Target Adverse Events Link with Immunologic and Clinical Outcomes
- Author
-
Petra Simon, Paul Lloyd-Evans, Emily Buxton, Stephen M. Thirdborough, Sarah Halford, Angelica Cazaly, Toni Weinschenk, Ann Mander, Jana Stasakova, Christian H. Ottensmeier, Alan Anthoney, Sally Clive, Ann O'Callaghan, Gareth J. Thomas, Andy King, Duncan I. Jodrell, Ceri Edwards, Freda K. Stevenson, Lindsey Chudley, Ugur Sahin, Andrew Bateman, and Katy J. McCann
- Subjects
Cytotoxicity, Immunologic ,Male ,Cancer Research ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Cancer Vaccines ,Article ,DNA vaccination ,03 medical and health sciences ,0302 clinical medicine ,Carcinoembryonic antigen ,Immune system ,Vaccines, DNA ,Medicine ,Humans ,Adverse effect ,biology ,business.industry ,Carcinoma ,Cancer ,medicine.disease ,Carcinoembryonic Antigen ,Vaccination ,Oncology ,Naked DNA ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Female ,Antibody ,business ,Oligopeptides ,030215 immunology - Abstract
Purpose: We have clinically evaluated a DNA fusion vaccine to target the HLA-A*0201–binding peptide CAP-1 from carcinoembryonic antigen (CEA605–613) linked to an immunostimulatory domain (DOM) from fragment C of tetanus toxin. Experimental Design: Twenty-seven patients with CEA-expressing carcinomas were recruited: 15 patients with measurable disease (arm-I) and 12 patients without radiological evidence of disease (arm-II). Six intramuscular vaccinations of naked DNA (1 mg/dose) were administered up to week 12. Clinical and immunologic follow-up was up to week 64 or clinical/radiological disease. Results: DOM-specific immune responses demonstrated successful vaccine delivery. All patients without measurable disease compared with 60% with advanced disease responded immunologically, while 58% and 20% expanded anti-CAP-1 CD8+ T cells, respectively. CAP-1–specific T cells were only detectable in the blood postvaccination but could also be identified in previously resected cancer tissue. The gastrointestinal adverse event diarrhea was reported by 48% of patients and linked to more frequent decreases in CEA (P < 0.001) and improved global immunologic responses [anti-DOM responses of greater magnitude (P < 0.001), frequency (P = 0.004), and duration] compared with patients without diarrhea. In advanced disease patients, decreases in CEA were associated with better overall survival (HR = 0.14, P = 0.017). CAP-1 peptide was detectable on MHC class I of normal bowel mucosa and primary colorectal cancer tissue by mass spectrometry, offering a mechanistic explanation for diarrhea through CD8+ T-cell attack. Conclusions: Our data suggest that DNA vaccination is able to overcome peripheral tolerance in normal and tumor tissue and warrants testing in combination studies, for example, by vaccinating in parallel to treatment with an anti-PD1 antibody. Clin Cancer Res; 22(19); 4827–36. ©2016 AACR.
- Published
- 2016
- Full Text
- View/download PDF
28. Immune Tolerance to Tumor Antigens Occurs in a Specialized Environment of the Spleen
- Author
-
Giacomo Desantis, Vincenzo Bronte, Steffen Walter, Mariacristina Chioda, Toni Weinschenk, Anna Cabrelle, Elisa Peranzoni, Susanna Mandruzzato, Jordi Ochando, and Stefano Ugel
- Subjects
Myeloid ,Cell Survival ,medicine.medical_treatment ,Lymphocyte ,Spleen ,Biology ,CD8-Positive T-Lymphocytes ,Monocyte ,LYMPHOCYTES ,Cancer Vaccines ,General Biochemistry, Genetics and Molecular Biology ,Monocytes ,Immune tolerance ,Tolerance ,Mice ,INFLAMMATORY MONOCYTES ,Antigen ,Antigens, Neoplasm ,Cell Line, Tumor ,Neoplasms ,CYCLOPHOSPHAMIDE ,medicine ,Immune Tolerance ,SUPPRESSOR-CELLS ,Animals ,Humans ,REGULATORY T-CELLS ,MYELOID CELLS ,ADOPTIVE IMMUNOTHERAPY ,lcsh:QH301-705.5 ,Cell Proliferation ,Peripheral tolerance ,Immunotherapy ,Marginal zone ,CANCER ,medicine.anatomical_structure ,PROGENITOR CELLS ,INFILTRATION ,lcsh:Biology (General) ,Immunology ,REGULATORY T-CELLS, SUPPRESSOR-CELLS, ADOPTIVE IMMUNOTHERAPY, INFLAMMATORY MONOCYTES, PROGENITOR CELLS, MYELOID CELLS, CANCER, CYCLOPHOSPHAMIDE, INFILTRATION, LYMPHOCYTES ,Immunologic Memory - Abstract
Peripheral tolerance to tumor antigens (Ags) is a major hurdle for antitumor immunity. Draining lymph nodes are considered the privileged sites for Ag presentation to T cells and for the onset of peripheral tolerance. Here, we show that the spleen is fundamentally important for tumor-induced tolerance. Splenectomy restores lymphocyte function and induces tumor regression when coupled with immunotherapy. Splenic CD11b(+)Gr-1(int)Ly6C(hi) cells, mostly comprising proliferating CCR2(+)-inflammatory monocytes with features of myeloid progenitors, expand in the marginal zone of the spleen. Here, they alter the normal tissue cytoarchitecture and closely associate with memory CD8(+) T cells, cross-presenting tumor Ags and causing their tolerization. Because of its high proliferative potential, this myeloid cell subset is also susceptible to low-dose chemotherapy, which can be exploited as an adjuvant to passive immunotherapy. CCL2 serum levels in cancer patients are directly related to the accumulation of immature myeloid cells and are predictive for overall survival in patients who develop a multipeptide response to cancer vaccines.
- Published
- 2012
29. Exploiting the glioblastoma peptidome to discover novel tumour-associated antigens for immunotherapy
- Author
-
Pierre-Yves Dietrich, Harpreet Singh-Jasuja, Jens Fritsche, Steffen Walter, Norbert Hilf, Judith Bucher, Katharina Dorsch, Christel Herold-Mende, Jennifer Lohr, Sylvia Flohr, Peter Lewandrowski, Stefan Stevanovic, Verona Vass, Paul R. Walker, Claudia Trautwein, Philipp Beckhove, Valérie Dutoit, Oliver Schoor, Toni Weinschenk, and Hans-Georg Rammensee
- Subjects
Antigens, CD31/metabolism ,Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism ,medicine.medical_treatment ,T cell ,Cell ,RNA, Messenger/metabolism ,Antigens, CD/metabolism ,Human leukocyte antigen ,CD8-Positive T-Lymphocytes ,Biology ,CD8-Positive T-Lymphocytes/immunology ,Mass Spectrometry ,Antigens, Neoplasm/chemistry/immunology/therapeutic use ,Antigen ,Antigens, CD ,Antigens, Neoplasm ,Sequence Analysis, Protein ,In vivo ,Glial Fibrillary Acidic Protein ,Glioblastoma/immunology/pathology/therapy ,medicine ,Humans ,RNA, Messenger ,Oligonucleotide Array Sequence Analysis ,ddc:616 ,Antigen Presentation ,Antigen Presentation/physiology ,HLA-A Antigens ,Brain Neoplasms ,Receptor-Like Protein Tyrosine Phosphatases, Class 5 ,Gene Expression Profiling ,Cytokines/metabolism ,Glial Fibrillary Acidic Protein/metabolism ,Immunotherapy ,Flow Cytometry ,Platelet Endothelial Cell Adhesion Molecule-1 ,medicine.anatomical_structure ,Immunology ,HLA-A Antigens/analysis/chemistry/immunology ,Peptides/analysis/immunology ,Cytokines ,Neurology (clinical) ,Glioblastoma ,Peptides ,CD8 ,Ex vivo ,Brain Neoplasms/immunology/pathology/therapy ,Chromatography, Liquid - Abstract
Peptides presented at the cell surface reflect the protein content of the cell; those on HLA class I molecules comprise the critical peptidome elements interacting with CD8 T lymphocytes. We hypothesize that peptidomes from ex vivo tumour samples encompass immunogenic tumour antigens. Here, we uncover >6000 HLA-bound peptides from HLA-A*02(+) glioblastoma, of which over 3000 were restricted by HLA-A*02. We prioritized in-depth investigation of 10 glioblastoma-associated antigens based on high expression in tumours, very low or absent expression in healthy tissues, implication in gliomagenesis and immunogenicity. Patients with glioblastoma showed no T cell tolerance to these peptides. Moreover, we demonstrated specific lysis of tumour cells by patients' CD8(+) T cells in vitro. In vivo, glioblastoma-specific CD8(+) T cells were present at the tumour site. Overall, our data show the physiological relevance of the peptidome approach and provide a critical advance for designing a rational glioblastoma immunotherapy. The peptides identified in our study are currently being tested as a multipeptide vaccine (IMA950) in patients with glioblastoma.
- Published
- 2012
30. Phase I adoptive cellular therapy trial with ex-vivo stimulated autologous CD8+ T-cells against multiple targets (ACTolog® IMA101) in patients with relapsed and/or refractory solid cancers
- Author
-
Toni Weinschenk, A. Stungis, R. Mendrzyk, A. Satelli, H. Ma, Patrick Hwu, K. Gharpure, Borje S. Andersson, J. Fritsche, Apostolia-Maria Tsimberidou, Oliver Schoor, Steffen Walter, C. Stewart, G. Stephens, Dominik Maurer, David Vining, Harpreet Singh, A. Mohamed, Carsten Reinhardt, and Cassian Yee
- Subjects
Cancer Research ,Oncology ,Refractory ,business.industry ,Cancer research ,Adoptive cellular therapy ,Medicine ,Cytotoxic T cell ,In patient ,Hematology ,business ,ACTolog IMA101 ,Ex vivo - Published
- 2018
31. Abstract CT127: Phase I adoptive cellular therapy trial with actively personalized, multi-targeted CD8+ T-cells in patients with relapsed and/or refractory solid cancers (ACTologIMA101-101)
- Author
-
Kerry Sieger, Cassian Yee, Apostolia-Marie Tsimberidou, Steffen Walter, Arun Satelli, Patrick Hwu, Norbert Hilf, Ali Mohamed, Dominik Maurer, Oliver Schoor, Yannick Bulliard, Borje S. Andersson, Sabrina Kuttruff, Carsten Reinhardt, Jens Fritsche, Toni Weinschenk, Geoffrey Stephens, Hong Ma, Chad Stewart, and Harpreet Singh
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,T cell ,medicine.medical_treatment ,Cancer ,Immunotherapy ,medicine.disease ,Clinical trial ,medicine.anatomical_structure ,Antigen ,Internal medicine ,medicine ,Cytotoxic T cell ,business ,CD8 ,Progressive disease - Abstract
Introduction: Adoptive cellular therapy (ACT), which includes the administration of autologous or allogenic anti-tumor T lymphocytes after ex vivo manipulation and expansion, has demonstrated substantial clinical benefit in some hematological cancer types. Particularly for solid cancers, only a relatively small proportion of patients has benefited from these advances due in part to i) lack of suitable immunotherapy targets with high specificity in solid cancers and ii) frequent relapse following immunotherapy to single targets often associated with loss of target expression in the tumor. The actively personalized IMA101 (ACTolog®) process, utilizing autologous antigen-specific T cells to multiple selected tumor targets, is intended to address these limitations. IMA101-101 is a first-in-human clinical trial in patients, with relapsed or refractory solid cancers, whose tumors express at least one target from a pre-defined warehouse of 8 cancer-specific targets. These targets were identified using Immatics' mass spectrometry (MS)-guided XPRESIDENT technology, and carefully selected for their highly tumor-associated expression profile. As such, ACTolog is a multi-target approach where up to four products with four different tumor-specificities will be infused per patient. Methods: One key defining feature of the approach used in this trial is the generation of robust T cells following a proprietary process, where autologous T cells are specifically primed against the expressed ACTolog targets in the presence of IL-21 followed by HLA tetramer-guided cell sorting and rapid expansion. This process has been shown to result in T-cell products with higher frequencies of central memory T cells, extended in vivo persistence and clinical response1,2. Patients are enrolled depending on their HLA-A*02:01 positivity and the expression of warehouse target(s) in the tumor. As the patients participating in this trial have a high unmet medical need (e.g. very poor prognosis and/or refractory or recurrent disease following multiple lines of therapy), treatment with IMA101 T cell products will take place when patients experience recurrence or progressive disease or when therapy is no longer warranted. It is planned that the first-in-human trial will enroll up to 20 patients. Results: Initial data from the patients enrolled in this ongoing Phase 1 clinical trial will be presented at the meeting with a focus on feasibility, safety, and T cell persistence and functionality. Conclusions: The actively personalized ACTolog® approach is unique in that it addresses the scarcity of suitable tumor antigens by using novel tumor targets identified via the MS-guided XPRESIDENT® technology. In addition, it is a multi-target approach intended to generate broad anti-tumor activity with decreased risk of tumor relapse due to loss of target expression. References: 1. Chapuis, AG, Desmarais, C, Emerson R, Schmit TM, Shibuya K, Lai, I, Wagener F, Chou J, Roberts IM, Coffey DG, Warren E, Robbins H, Greenberg PD, Yee C (2017). "Tracking the fate and origin of clinically relevant adoptively transferred CD8+ T cells in vivo." Science Immunology 2(9) 2. Yee C. “The use of endogenous T cells for adoptive transfer.” Immunol Rev. Jan;257(1):250-63. 2014 Citation Format: Hong Ma, Sabrina Kuttruff, Chad Stewart, Yannick Bulliard, Geoffrey Stephens, Oliver Schoor, Arun Satelli, Norbert Hilf, Kerry Sieger, Jens Fritsche, Dominik Maurer, Ali Mohamed, Toni Weinschenk, Carsten Reinhardt, Steffen Walter, Harpreet Singh, Patrick Hwu, Cassian Yee, Borje Andersson, Apostolia-Marie Tsimberidou. Phase I adoptive cellular therapy trial with actively personalized, multi-targeted CD8+ T-cells in patients with relapsed and/or refractory solid cancers (ACTologIMA101-101) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr CT127.
- Published
- 2018
32. Abstract 2789: Development of highly potent T-cell receptor bispecifics with picomolar activity against tumor-specific HLA ligands
- Author
-
Toni Weinschenk, Leonie Alten, Meike Hutt, Carsten Reinhardt, Harpreet Singh-Jasuja, Jens Fritsche, Sebastian Bunk, Dominik Maurer, Felix Unverdorben, Claudia Wagner, Oliver Schoor, Martin Hofmann, and Mathias Ferber
- Subjects
Cancer Research ,medicine.diagnostic_test ,biology ,Chemistry ,Effector ,T-cell receptor ,Human leukocyte antigen ,Cell sorting ,Fusion protein ,Flow cytometry ,Cell biology ,Oncology ,Antigen ,medicine ,biology.protein ,Antibody - Abstract
T-cell receptor (TCR)-based immunotherapy has emerged as a promising treatment modality for malignant diseases. TCRs naturally recognize human leucocyte antigen (HLA)-bound peptides from foreign and endogenous antigens regardless of their source proteins' extracellular or intracellular location. Using its proprietary discovery engine XPRESIDENT®, Immatics can identify, quantify, and validate Tumor-Associated Peptides (TUMAPs) exclusively presented in cancer tissues. Immatics has established state-of-the-art technology to discover and affinity maturate TUMAP-specific TCRs originating from human T-cell repertoire. The maturated single-chain TCRs (scTv) are used to build a pipeline of highly potent T-cell engaging bispecific TCR molecules directed against TUMAPs. In brief, we use artificial antigen-presenting cells to selectively expand TUMAP-specific T-cells from which the coding TCR sequence is retrieved by 5'RACE after highly sensitive flow cytometry-based single cell sorting. About 50-150 TCRs per TUMAP are transiently re-expressed on human T-cells and extensively characterized for their functional properties. TCRs exhibiting highly active and specific TUMAP recognition are selected for yeast surface display to generate stabilized and affinity maturated scTv. The maturated scTv are engineered into Immatics' proprietary bispecific TCR scaffold comprising a humanized T-cell recruiting antibody domain for potent redirection and activation of T-cells against TUMAPs and an effector function-silenced IgG1 Fc domain. Here we present data of our bispecific TCR program targeting the TUMAP Ag008-01 bound to HLA-A*02. We confirmed the abundant presence of Ag008-01 in several cancer indications and its absence in human normal tissues by using XPRESIDENT® technology combining quantitative mass spectrometry, transcriptomics and bioinformatics. Based on the parental TCR showing highly active and specific recognition of Ag008-01, we generated stabilized and affinity maturated scTv variants with significant improvement in binding affinity towards Ag008-01 in the range of 1000-fold and higher. The maturated scTv variants showed no or minimal binding to off-target peptides selected from the XPRESIDENT® normal tissue database based on the criteria of highest sequence similarity to Ag008-01. By incorporating the maturated scTv into our proprietary bispecific TCR format, which outperformed five alternative TCR bispecific format designs, we obtained highly potent bispecific TCR molecules with picomolar activity. We observed half-maximal lysis of Ag008-01 expressing tumor cell lines at TCR bispecific concentrations below 100 picomolar while no reactivity was observed towards a panel of cell lines lacking Ag008-01 expression. Our data support proof-of-concept for the design of our novel class of T-cell engaging bispecific TCR-antibody fusion proteins. Citation Format: Sebastian Bunk, Martin Hofmann, Felix Unverdorben, Leonie Alten, Meike Hutt, Claudia Wagner, Oliver Schoor, Mathias Ferber, Jens Fritsche, Toni Weinschenk, Harpreet Singh-Jasuja, Dominik Maurer, Carsten Reinhardt. Development of highly potent T-cell receptor bispecifics with picomolar activity against tumor-specific HLA ligands [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2789.
- Published
- 2018
33. Abstract 3588: ACTallo: A novel approach using gamma-delta T cells to allogeneic cellular therapy to treat cancer
- Author
-
Ali Mohamed, Yannick Bulliard, Melinda Mata, Mo Dao, Leonie Alten, Aleksandra Nowicka, Sabrina Kuttruff, Toni Weinschenk, Steffen Walter, and Sarah Missel
- Subjects
Cancer Research ,LAG3 ,medicine.medical_treatment ,T-cell receptor ,Cancer ,Endogeny ,Context (language use) ,Biology ,medicine.disease ,Cell therapy ,Oncology ,Cancer immunotherapy ,medicine ,Cancer research ,CD8 - Abstract
Adoptive cellular therapy (ACT) has dramatically changed the landscape of cancer immunotherapy; however, the complex manufacture of individualized products from late-stage, heavily pre-treated cancer patients, together with high cost-of-goods, encourages the development of simpler ‘off-the-shelf' alternatives. The unique properties of γδ (gamma-delta) T cells make them a seductive candidate for effective allogeneic ACT. In contrast to αβ T cells, γδ T cells are endowed with intrinsic antitumor activity while unable to induce graft-versus-host disease. γδ T cells have a high proliferative potential before exhibiting exhaustion. Moreover, in the context of αβ TCR-engineered ACT, γδ T cells pose a lesser risk of off-target toxicity, due to the absence of expression of endogenous αβ TCR and hence, risk of mispairing with the engineered αβ TCR. Here, we will present the ACTallo® platform, which combines the unique properties of γδ T cells with Immatics' XPRESIDENT® target/TCR discovery platform. The ACTallo® process selectively expands Vγ9Vδ2 T cells that are retrovirally-transduced to co-express CD8 together with a tumor-specific αβ TCR. Over a period of 45 days or less, ACTallo® γδ T cells can expand >30,000 fold. γδ T cells obtained this way recognize tumor cells, express cytokines, degranulate and kill tumor cells specifically. In addition, ACTallo® γδ T cells show no signs of exhaustion after expansion, as evidenced by the lack of expression of PD1, TIM3 or LAG3. ACTallo® is intended to overcome the limitation of autologous cellular therapies towards off-the-shelf approach, by combining the large-scale expansion potential of allogeneic γδ T cells with novel TCRs against XPRESIDENT® mass-spectrometry guided tumor-specific targets. Citation Format: Mo Dao, Melinda Mata, Leonie Alten, Aleksandra Nowicka, Sabrina Kuttruff, Sarah Missel, Ali Mohamed, Toni Weinschenk, Yannick Bulliard, Steffen Walter. ACTallo: A novel approach using gamma-delta T cells to allogeneic cellular therapy to treat cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3588.
- Published
- 2018
34. Front Cover: Translating Immunopeptidomics to Immunotherapy-Decision-Making for Patient and Personalized Target Selection
- Author
-
Jens Fritsche, Barbara Rakitsch, Franziska Hoffgaard, Michael Römer, Heiko Schuster, Daniel J. Kowalewski, Martin Priemer, Vlatka Stos-Zweifel, Helen Hörzer, Arun Satelli, Annika Sonntag, Valentina Goldfinger, Colette Song, Andrea Mahr, Martina Ott, Oliver Schoor, and Toni Weinschenk
- Subjects
Molecular Biology ,Biochemistry - Published
- 2018
35. HepaVac-101 first-in-man therapeutic cancer vaccine phase I/II clinical trial for hepatocellular carcinoma patients
- Author
-
Joerg Ludwig, Regina Heidenreich, Hans-Georg Rammensee, Markus W. Loeffler, Toni Weinschenk, Roberto S. Accolla, Sven Francque, Yuk Ting Ma, Tanguy Chaumette, Andrea Mayer-Mokler, Senta Ulrike Gnad-Vogt, Luigi Buonaguro, Ester Simeone, Christian Flohr, Carsten Reinhardt, Alfred Koenigsrainer, Mercedes Iñarrairaegui, Cécile Gouttefangeas, Harpreet Singh, and Antonio Avallone
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Cancer ,medicine.disease ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Phase i ii ,030220 oncology & carcinogenesis ,Internal medicine ,Hepatocellular carcinoma ,medicine ,Cancer vaccine ,business ,Survival rate ,Cause of death - Abstract
TPS3135Background: HCC is the third leading cause of death from cancer globally with an extremely variable 5-year survival rate. The HepaVac-101 phase I/II, first-in-man, therapeutic cancer vaccine...
- Published
- 2018
36. Translating Immunopeptidomics to Immunotherapy-Decision-Making for Patient and Personalized Target Selection
- Author
-
Annika Sonntag, Toni Weinschenk, Michael Römer, Andrea Mahr, Arun Satelli, Martin Priemer, Daniel J. Kowalewski, Jens Fritsche, Franziska Hoffgaard, Vlatka Stos-Zweifel, Colette Song, Helen Hörzer, Heiko Schuster, Oliver Schoor, Martina Ott, Valentina Goldfinger, and Barbara Rakitsch
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Decision Making ,Human leukocyte antigen ,Computational biology ,Immunopeptidome ,Biochemistry ,Mass Spectrometry ,Epitopes ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Antigens, Neoplasm ,HLA Antigens ,Neoplasms ,medicine ,Humans ,RNA, Messenger ,Molecular Biology ,Research Articles ,Selection (genetic algorithm) ,Proteogenomics ,Antigen Presentation ,business.industry ,Precision medicine ,High-Throughput Nucleotide Sequencing ,Immunotherapy ,Peptide Fragments ,Treatment efficacy ,Clinical trial ,Label-free quantification ,030104 developmental biology ,030220 oncology & carcinogenesis ,Label‐free quantitation ,business ,Research Article - Abstract
Immunotherapy is revolutionizing cancer treatment and has shown success in particular for tumors with a high mutational load. These effects have been linked to neoantigens derived from patient‐specific mutations. To expand efficacious immunotherapy approaches to the vast majority of tumor types and patient populations carrying only a few mutations and maybe not a single presented neoepitope, it is necessary to expand the target space to non‐mutated cancer‐associated antigens. Mass spectrometry enables the direct and unbiased discovery and selection of tumor‐specific human leukocyte antigen (HLA) peptides that can be used to define targets for immunotherapy. Combining these targets into a warehouse allows for multi‐target therapy and accelerated clinical application. For precise personalization aimed at optimally ensuring treatment efficacy and safety, it is necessary to assess the presence of the target on each individual patient's tumor. Here we show how LC‐MS paired with gene expression data was used to define mRNA biomarkers currently being used as diagnostic test IMADETECT™ for patient inclusion and personalized target selection within two clinical trials (NCT02876510, NCT03247309). Thus, we present a way how to translate HLA peptide presentation into gene expression thresholds for companion diagnostics in immunotherapy considering the peptide‐specific correlation to its encoding mRNA.
- Published
- 2018
37. Phase I trial evaluating genetically modified autologous T cells (ACTengine IMA201) expressing a T-cell receptor recognizing a cancer/germline antigen in patients with squamous NSCLC or HNSCC
- Author
-
Partow Kebriaei, Patrick Hwu, Harpreet Singh, Agathe Bourgogne, Hong Ma, Carsten Reinhardt, George R. Blumenschein, Toni Weinschenk, and Steffen Walter
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,medicine.medical_treatment ,T-cell receptor ,Cancer ,Target peptide ,Immunotherapy ,medicine.disease ,Germline ,Genetically modified organism ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Antigen ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Receptor ,business - Abstract
TPS78 Background: Adoptive cellular therapy (ACT) has dramatically changed the landscape of immunotherapy. T-cell receptor (TCR) engineered ACT approach has contributed to success in solid tumors. ACTengine IMA201 is a product based on autologous T cells engineered to express a naturally occurring TCR specific to a cancer-germline peptide. The target peptide has been characterized in depth by Immatics’ proprietary antigen discovery platform, XPRESIDENT showing exceptional specificity, copy number and expression homogeneity. XPRESIDENT has applied two independent methodologies to confirm the selectivity of tumor target: i) ultra-sensitive quantitative immunopeptidome analyses by mass spectrometry and ii) quantitative mRNA expression analyses by RNASeq. Immatics´ TCR discovery platform is optimized to identify TCRs with low micromolar affinity, specific recognition of tumor cell lines and lack of recognition of healthy normal cells. Methods: This study is an open-label first-in-human dose-escalating phase I trial investigating safety, tolerability and clinical activity in end-stage solid tumor patients. Key eligibility criteria include: recurrent or refractory squamous non-small cell lung cancer or head and neck squamous cell cancer, HLA-A*02:01 phenotype, qPCR biomarker positive from a tumor biopsy, prior established lines of therapy, RECIST v1.1 measurable lesions and ECOG score 0 or 1. At baseline, patients will undergo leukapheresis to collect mononuclear cells (PBMC). The activated PBMC will be transduced with a lentiviral vector for manufacturing of IMA201. IMA201 is infused after a pre-conditioning (lymphodepletion) followed by LD-IL2. The primary objective is to assess safety and to identify the maximum tolerated dose. Secondary endpoints include overall response rate (RECIST and irRC), PFS and OS. The translational objectives include: i) the persistence and functionality of IMA201 in vivo, ii) correlative biomarkers for clinical success, and iii) target expression levels in the tumor. Enrollment to the study is currently ongoing. Clinical trial information: NCT03247309 .
- Published
- 2018
38. Phase I adoptive cellular therapy trial with ex-vivo stimulated autologous CD8+ T-cells against multiple targets (ACTolog IMA101) in patients with relapsed and/or refractory solid cancers
- Author
-
Carsten Reinhardt, Patrick Hwu, Cassian Yee, Harpreet Singh, Amir A. Jazaeri, Steffen Walter, Apostolia Maria Tsimberidou, Borje S. Andersson, Toni Weinschenk, Chad Stewart, and Hong Ma
- Subjects
Cancer Research ,business.industry ,medicine.medical_treatment ,Immunotherapy ,Tumor antigen ,Oncology ,Antigen ,Tumor Escape ,Refractory ,medicine ,Cancer research ,Cytotoxic T cell ,In patient ,business ,Ex vivo - Abstract
TPS77 Background: Adoptive cellular therapy (ACT) has dramatically changed the landscape of immunotherapy; however, only a small proportion of solid tumor patients have benefited from these advances due to i) heterogeneity of tumor antigen expression, ii) tumor escape (e.g. only one target is addressed), or iii) off-target toxicities (e.g. expression of targets on normal tissues). The ACTolog concept, utilizing antigen specific T cells (IMA101) against targets identified by the Immatics’ proprietary XPRESIDENT technology, is intended to overcome these limitations by addressing multiple novel relevant tumor antigens per patient. ACTolog is a personalized, multi-targeted ACT approach in which autologous T-cell products are manufactured against the most relevant tumor target peptides for individual patients whose tumors are positive against a predefined target warehouse. Methods: This study is an open-label first-in-human phase I trial in patients with relapsed or refractory solid tumors expressing at least one target from a warehouse of 8 cancer targets. Key eligibility criteria include: HLA-A*02:01 phenotype, qPCR expression of warehouse target(s), prior established lines of therapy, RECIST v1.1 measurable lesions, and ECOG performance status 0 or 1. At baseline, patients will undergo leukapheresis to collect mononuclear cells for manufacturing of IMA101 cells. Patients will receive their last line of established therapy during the production phase of IMA101. IMA101 will be infused after a pre-conditioning regimen (lymphodepletion) followed by LD-IL2. The primary objective is to assess safety and tolerability of IMA101. Secondary endpoints include overall response rate (RECIST and irRC), PFS and OS. The translational objective is to assess the in vivo persistence and ex vivo functionality of transferred T cells in addition to evaluation of target expression in tumors. Enrollment to the study is currently ongoing. Clinical trial information: NCT02876510 .
- Published
- 2018
39. Sorafenib, but not sunitinib, affects function of dendritic cells and induction of primary immune responses
- Author
-
Peter Brossart, Markus P. Radsak, Harpreet Singh-Jasuja, Daniela Werth, Toni Weinschenk, Madeleine M. Hipp, Steffen Walter, Norbert Hilf, and Katharina M. Brauer
- Subjects
Niacinamide ,Sorafenib ,Indoles ,Pyridines ,Immunology ,Antineoplastic Agents ,Apoptosis ,CD8-Positive T-Lymphocytes ,Pharmacology ,Biology ,urologic and male genital diseases ,Major histocompatibility complex ,T-Lymphocytes, Regulatory ,Biochemistry ,Peripheral blood mononuclear cell ,Mice ,Immune system ,Cell Movement ,In vivo ,Sunitinib ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Pyrroles ,Cells, Cultured ,Phenylurea Compounds ,Benzenesulfonates ,Granulocyte-Macrophage Colony-Stimulating Factor ,Dextrans ,Dendritic Cells ,Cell Biology ,Hematology ,Endocytosis ,female genital diseases and pregnancy complications ,Mice, Inbred C57BL ,Toll-Like Receptor 4 ,biology.protein ,Cytokines ,Female ,Interleukin-4 ,Lymphocyte Culture Test, Mixed ,Tyrosine kinase ,Cell Division ,Signal Transduction ,medicine.drug - Abstract
The tyrosine kinase inhibitors sorafenib and sunitinib are approved for the treatment of patients with malignant diseases. To analyze the possible use of these compounds in combination with immunotherapeutic approaches, we analyzed the effects of both inhibitors on the immunostimulatory capacity of human dendritic cells (DCs) and the induction of primary immune responses in vivo. Sorafenib, but not sunitinib, inhibits function of DCs, characterized by reduced secretion of cytokines and expression of CD1a, major histocompatibility complex, and costimulatory molecules in response to TLR ligands as well as by their impaired ability to migrate and stimulate T-cell responses. These inhibitory effects are mediated by inhibition of PI3 and MAP kinases and NFκB signaling. In contrast, sorafenib had no influence on the phenotype and proliferation of T cells. To analyze the effects of both TKIs on cytotoxic T-cell induction in vivo, C57BL/6 mice were pretreated with sorafenib or sunitinib and immunized with OVA257-264 peptide. Sorafenib, but not sunitinib, application significantly reduced the induction of antigen-specific T cells. Numbers of regulatory T cells were reduced in peripheral blood mononuclear cells from mice treated with sunitinib. These results indicate that sunitinib, but not sorafenib, is suitable for combination with immunotherapeutic approaches for treatment of cancer patients.
- Published
- 2008
40. siRNA binding proteins of microglial cells: PKR is an unanticipated ligand
- Author
-
Toni Weinschenk, Ketai Guo, Hermann J. Schluesener, and Zhiren Zhang
- Subjects
Small interfering RNA ,SiRNA binding ,Trans-acting siRNA ,DNA, Single-Stranded ,Biology ,Ligands ,Biochemistry ,Mice ,eIF-2 Kinase ,RNA interference ,Animals ,Humans ,Gene silencing ,Gene Silencing ,RNA, Small Interfering ,Molecular Biology ,Cells, Cultured ,Endothelial Cells ,RNA ,Cell Biology ,Transfection ,Molecular biology ,RNA silencing ,Microscopy, Fluorescence ,Microglia ,Carrier Proteins - Abstract
Small interfering RNA (siRNA), double-stranded RNA (dsRNA) 21-23 nucleotides (nt) long with two nt 3' overhangs, has been shown to mediate powerful sequence-specific gene silence in mammalian cells through RNA interference (RNAi). Due to its high efficiency and high specificity siRNA has been used as a powerful post genomic tool and a potent therapeutic candidate. However, there is still a lot to learn about the mobility of siRNA inside cells and the cellular factors that might interfere with the specificity and activity of siRNA. Microglia are the brain's effector cells of the innate immune system and suitable targets in the development of novel therapeutic strategies. Here, we show the cellular uptake and intracellular distribution of siRNA in murine microglial N9 cells. siRNA was internalized by microglial N9 cells without transfection reagent and mainly localized to the endosomes However, no significant gene silencing effects were observed. Its cellular uptake and cellular distribution pattern were similar with that of a same length single stranded DNA (ssDNA). Further, cellular binding proteins of siRNA were purified and identified by mass spectrometry. Negative control siRNA and siRNA targeted to beta-actin were used in this part of experiment. Most of the siRNA binding proteins for negative control siRNA and siRNA targeted to beta-actin were dsRNA-binding proteins, such as dsRNA-dependent protein kinase R (PKR). Furthermore, both control siRNA and siRNA targeted to beta-actin activated PKR in N9 cells, which suggest that siRNA might cause off-target effects through activation of PKR.
- Published
- 2006
41. MAGED4 – expression in renal cell carcinoma and identification of an HLA-A*25-restricted MHC class I ligand from solid tumor tissue
- Author
-
Tobias Krüger, Toni Weinschenk, Christian Reichle, Oliver Schoor, Hans-Georg Rammensee, Stefan Stevanovic, Björn F. Krämer, Arnulf Stenzl, Jörg Hennenlotter, and Margaret Müller
- Subjects
Cancer Research ,Genes, MHC Class I ,Human leukocyte antigen ,Ligands ,urologic and male genital diseases ,Epitope ,Antigen ,Antigens, Neoplasm ,HLA Antigens ,Renal cell carcinoma ,MHC class I ,medicine ,Humans ,Carcinoma, Renal Cell ,neoplasms ,Pharmacology ,biology ,medicine.disease ,Molecular biology ,Kidney Neoplasms ,female genital diseases and pregnancy complications ,Neoplasm Proteins ,HLA-A ,Gene expression profiling ,Oncology ,Case-Control Studies ,biology.protein ,Molecular Medicine ,Cancer/testis antigens ,Peptides - Abstract
MAGE derived HLA ligands have repeatedly been shown to elicit T-cell responses against tumor cells. In renal cell carcinoma (RCC), however, only few T-cell epitopes from cancer testis antigens have been described. To identify potential candidates, we applied a combined approach of microarray/qPCR expression analysis and sequencing of HLA ligands from RCC by mass spectrometry. We analyzed the expression of 21 MAGE genes in ten RCC samples and two glioblastoma samples and could identify the first MHC class I ligand NIGDEALIGRW from MAGED4 presented by HLA-A*25 on RCC solid tumor tissue. MAGED4 was expressed in 30% of RCC and both glioblastoma samples. Among the other MAGE family members only MAGEB2 and -C1 and the broadly expressed MAGED1, -D2, -F1 and -H1 were expressed in RCC. Ligands from MAGED4 could thus be interesting tumor-associated antigens in a subset of RCC, even though the identified ligand is presented by a rather rare allele.
- Published
- 2005
42. Effects of Imatinib on Monocyte-Derived Dendritic Cells Are Mediated by Inhibition of Nuclear Factor-κB and Akt Signaling Pathways
- Author
-
Frank Grünebach, Silke Appel, Tim H. Brümmendorf, Anette Rupf, Markus M. Weck, Oliver Schoor, Peter Brossart, and Toni Weinschenk
- Subjects
Cancer Research ,Lipopolysaccharide Receptors ,Antineoplastic Agents ,Dendritic cell differentiation ,Protein Serine-Threonine Kinases ,Biology ,Monocytes ,Piperazines ,Growth factor receptor ,Proto-Oncogene Proteins ,medicine ,Humans ,Protein kinase B ,Immunosuppression Therapy ,Gene Expression Profiling ,Monocyte ,NF-kappa B ,Cell Differentiation ,Dendritic Cells ,Dendritic cell ,Cell biology ,Pyrimidines ,Imatinib mesylate ,medicine.anatomical_structure ,Oncology ,Benzamides ,Imatinib Mesylate ,Cancer research ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Tyrosine kinase ,Signal Transduction - Abstract
Dendritic cells are the most powerful antigen-presenting cells playing a decisive role for the initiation and maintenance of primary immune responses. However, signaling pathways involved in the differentiation of these cells have not been fully determined. Imatinib is a novel tyrosine kinase inhibitor effective against Abl kinases, c-Kit, and platelet-derived growth factor receptor. Using this compound, we show that human monocyte-derived dendritic cells generated in the presence of therapeutic concentrations of imatinib show a reduced expression of CD1a, MHC class I and II, and costimulatory molecules as well as decreased secretion of chemokines and cytokines resulting in an impaired capacity of dendritic cells to elicit primary T-cell responses. Using Western blot analyses, we found that these effects are mediated by inhibition of phosphatidylinositol 3-kinase/Akt pathways and a pronounced down-regulation of nuclear localized protein levels of nuclear factor-κB family members. Importantly, using blocking antibodies and tyrosine kinase inhibitors, we show that the inhibitory effects of imatinib on dendritic cell differentiation are not mediated via platelet-derived growth factor receptor and c-Kit. Taken together, our study reveals that imatinib inhibits dendritic cell differentiation and function via Akt and nuclear factor-κB signal transduction. Importantly, we show that imatinib can inhibit the function of normal, nonmalignant cells that may result in immunosuppression of these patients.
- Published
- 2005
43. Uptake, intracellular distribution, and novel binding proteins of immunostimulatory CpG oligodeoxynucleotides in microglial cells
- Author
-
Toni Weinschenk, Zhiren Zhang, and Hermann J. Schluesener
- Subjects
Intracellular Fluid ,CpG Oligodeoxynucleotide ,Immunology ,Plasma protein binding ,Biology ,Nitric Oxide ,Binding, Competitive ,DNA-binding protein ,Mice ,chemistry.chemical_compound ,Adjuvants, Immunologic ,Polysaccharides ,Heterogeneous-Nuclear Ribonucleoprotein Group A-B ,Animals ,Immunology and Allergy ,Cells, Cultured ,Cellular localization ,Pyruvate Carboxylase ,ADP Ribose Transferases ,Innate immune system ,Oligonucleotide ,Dextran Sulfate ,hemic and immune systems ,DNA ,Thionucleotides ,respiratory system ,Microscopy, Fluorescence ,Oligodeoxyribonucleotides ,Neurology ,CpG site ,chemistry ,Biochemistry ,CpG Islands ,Microglia ,Neurology (clinical) ,Carrier Proteins ,Fluorescein-5-isothiocyanate ,Protein Binding - Abstract
Microglial cells are central components of the innate immune system of the brain and contribute to inflammatory and degenerative processes. DNA with unmethylated CpG dinucleotides is a potent stimulant of microglial cells. We have analyzed uptake, intracellular distribution, and cellular binding proteins of CpG oligdeoxynucleotides (ODNs) by the microglial cell line N9. The uptake of CpG-ODN is concentration-, time-, and temperature-dependent, but, interestingly, independent of the CpG dinucleotides. After internalisation, CpG-ODN localized to the cytoplasm and showed a typical speckled distribution pattern. We further purified the cellular binding proteins of CpG-ODN and identified several binding proteins by tryptic digestion and mass spectrometry. Most of the CpG-ODN binding proteins are RNA processing enzymes, which are important for RNA splicing, export, and stability. Further, we identified a protein, pigpen, which has not been observed in microglial cells, so far. These proteins apparently bind CpG-ODN with low selectivity, as binding is independent of CpG dinucleotides. Interference of immunostimulatory and therapeutic oligonucleotides with proteins and enzymes of RNA transport and processing has not been described so far and might affect the physiological functions of these proteins and also might influence cellular localization of therapeutic ODN. These findings are helpful in understanding the cellular fate of ODN and the nonsequence-specific effects of ODN and for rational design and evaluation of ODN-based therapeutic strategies.
- Published
- 2005
44. CD8+ T-cell response against MUC1-derived peptides in gastrointestinal cancer survivors
- Author
-
Karin Keller-Matschke, Horst-Dieter Becker, Toni Weinschenk, Cécile Gouttefangeas, Thomas Kratt, Stefan Stevanovic, Jasmin Dittmann, Hans-Georg Rammensee, and Tobias Heck
- Subjects
Male ,Cancer Research ,medicine.medical_treatment ,Immunology ,CD8-Positive T-Lymphocytes ,Epitope ,Epitopes ,Immune system ,Antigen ,Stomach Neoplasms ,Humans ,Immunology and Allergy ,Medicine ,Cytotoxic T cell ,Survivors ,Gastrointestinal cancer ,neoplasms ,MUC1 ,Aged ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Mucin-1 ,Immunotherapy ,Middle Aged ,medicine.disease ,Oncology ,Antibody Formation ,Female ,Colorectal Neoplasms ,business ,CD8 - Abstract
The tumor-associated antigens CEA, MUC1 and Her2/neu are broadly expressed in gastrointestinal tumors, and are attractive candidates for targeting by T-cell-based immunotherapy. However, little is known about the natural cytotoxic T-cell response of patients suffering from colorectal or gastric carcinoma against these three as well as other antigens. Using a quantitative reverse transcription-polymerase chain reaction-based assay for IFN-gamma, we analyzed the CD8+ T-cell repertoire present in the blood of HLA-A2+ gastrointestinal tumor survivors against five known epitopes derived from CEA, MUC1 and Her2/neu. The results show that most of the patients (16 from 22 tested) have detectable, peripheral CD8+ T cells directed against at least one of these three proteins. Interestingly, the majority of these patients reacts to the two MUC1-derived HLA-A*0201 epitopes tested (14 from 16), demonstrating that this protein represents one dominant target for CD8+ T cells in gastrointestinal cancer.
- Published
- 2004
45. A conserved sequence in the mouse variable T cell receptorα recombination signal sequence 23-bp spacer can affect recombination
- Author
-
Stefan Tenzer, Adrien Six, Jochen Probst, Steve Pascolo, Sibylle G. Blumenthal, Jürgen Dittmer, Oliver Schoor, Hans-Georg Rammensee, and Toni Weinschenk
- Subjects
Recombination, Genetic ,Base Sequence ,Immunology ,T-cell receptor ,Receptors, Antigen, T-Cell ,Locus (genetics) ,Thymus Gland ,Biology ,Molecular biology ,Conserved sequence ,Mice ,Plasmid ,T-Lymphocyte Subsets ,Mutation ,Animals ,Immunology and Allergy ,Recombination signal sequences ,Gene ,Conserved Sequence ,Recombination ,Palindromic sequence - Abstract
Although the V-gene segments coding for the TCR alpha and delta chains are mixed together in the alpha delta locus and are recombined by the same processes, some gene segments (TRAV) are rearranged only with TCR Jalpha gene segments, some (TRDV) only with TCR Ddelta gene segments and some (TRADV) with both. To date, no molecular signal is known that can characterize these three different types of gene segments. Studying the recombination signal sequences (RSS) of all mouse TCR V-gene segments we observed that 80% of the TRAV contain a palindrome sequence (CTGCAG) or its related variant CTGTAG in their 23-bp spacer. Using gel-shift assays we show that these sequences are specifically recognized by some nuclear proteins that are expressed by fresh thymocytes, fresh lymphocytes and tumor cells. Recombination assays on plasmid substrates in a pre-B cell line showed that RSS containing the CTGCAG sequence can impair recombination. From the protein fractions containing the CTGCAG-binding activity, three proteins were identified: G3BP1 (a nucleic-acid-binding protein with a proposed helicase activity) and two proteins from the high-mobility group (HMG) family--HMGB2 and HMGB3. We hypothesize that these proteins can affect recombination at the TCR alpha delta locus.
- Published
- 2004
46. Moderate degradation does not preclude microarray analysis of small amounts of RNA
- Author
-
S. Corvin, Toni Weinschenk, Jörg Hennenlotter, Hans-Georg Rammensee, Arnulf Stenzl, Stefan Stevanovic, and Oliver Schoor
- Subjects
Microarray analysis techniques ,Gene Expression Profiling ,Microchemistry ,Genetic Variation ,Reproducibility of Results ,RNA ,Ribosomal RNA ,Biology ,Nucleic Acid Denaturation ,Sensitivity and Specificity ,Molecular biology ,Kidney Neoplasms ,General Biochemistry, Genetics and Molecular Biology ,Specimen Handling ,Gene expression profiling ,Sample Size ,Gene expression ,Humans ,DNA microarray ,Carcinoma, Renal Cell ,Gene ,Oligonucleotide Array Sequence Analysis ,Biotechnology ,Laser capture microdissection - Abstract
Gene expression analysis by microarrays using small amounts of RNA is becoming more and more popular against the background of advances and increasing importance of small-sample acquisition methods like laser microdissection techniques. The quality of RNA preparations from such samples constitutes a frequent issue in this context. The aim of this study was to assess the impact of different extents of RNA degradation on the expression profile of the samples. We induced RNA degradation in human tumor and healthy tissue samples by endogeneous ribonucleases. Next, we amplified 20 ng total RNA degraded to different extents by two rounds of in vitro transcription and analyzed them using Affymetrix oligonucleotide microarrays. Expression differences for some genes were independently confirmed by real-time quantitative PCR. Our results suggest that gene expression profiles obtained from partially degraded RNA samples with still visible ribosomal bands exhibit a high degree of similarity compared to intact samples and that RNA samples of suboptimal quality might therefore still lead to meaningful results if used carefully.
- Published
- 2003
47. The allograft inflammatory factor-1 in Creutzfeldt-Jakob disease brains
- Author
-
Toni Weinschenk, Richard Meyermann, Martin H. Deininger, and H. J. Schluesener
- Subjects
education.field_of_study ,Pathology ,medicine.medical_specialty ,Histology ,Human leukocyte antigen ,Biology ,Molecular biology ,Reverse transcriptase ,Pathology and Forensic Medicine ,law.invention ,Blot ,Neurology ,law ,In vivo ,Physiology (medical) ,Recombinant DNA ,Allograft inflammatory factor 1 ,medicine ,cardiovascular diseases ,Neurology (clinical) ,biological phenomena, cell phenomena, and immunity ,education ,Cell activation ,Polymerase chain reaction - Abstract
The allograft inflammatory factor-1 (AIF-1) is a 17-kDa IFN-γ inducible Ca 2+ -binding EF-hadn protein that is encoded within the HLA class III genomic region and is involved in immune dysfunction and smooth muscle cell activation. We used immuncthistochemistry double labelling experiments to analyse the spatial distribution and cell-type-specific localisation of AIF-1 in the brains of patients who died as a result of sporadic Creutzfeldt-Jakob disease (CJD) and neuropathologically unaltered controls, Significantly more AIF-1 immunoreactive macrophages/ microglial cells and, interestingly, neurones were observed in CJD patients compared to controls. Western blotting confirmed more prominent AIF-1 immunoreactive bands of approximately 50 kDa in four CJD patients compared to three controls. Chaotropic SDS-PAGE of the recombinant AIF- resulted in almost complete reduction of the 50 kDa band and mass spectrometry revealed only AIF-1-specific tryptic protein fragments suggesting that trimerized AIF-1 is the predominant form in vivo. Finally, we analysed mechanisms of neuronal AIF-1 induction. Following H 2 O 2 challenge, a model of general cell stress, we observed the gradual induction of AIF-1 and, more interestingly, release to the supernatant of SKNSH neurones. Parallel reverse transcriptase polymerase chain reaction and sequencing was used to confirm AIF-1 mRNA expression.
- Published
- 2003
48. Tumor-associated antigen profiling in breast and ovarian cancer: mRNA, protein or T cell recognition?
- Author
-
C. Rentzsch, Toni Weinschenk, Diethelm Wallwiener, S. Kayser, Iris Watermann, and B. Gückel
- Subjects
Cancer Research ,T-Lymphocytes ,T cell ,Breast Neoplasms ,Major histocompatibility complex ,Immunophenotyping ,Immune system ,Antigen ,Antigens, Neoplasm ,Cell Line, Tumor ,MHC class I ,medicine ,Humans ,RNA, Messenger ,Ovarian Neoplasms ,Base Sequence ,biology ,Antigen processing ,Carcinoma ,Histocompatibility Antigens Class I ,Histocompatibility Antigens Class II ,General Medicine ,Transporter associated with antigen processing ,Neoplasm Proteins ,Gene expression profiling ,medicine.anatomical_structure ,Oncology ,Immunology ,biology.protein ,ATP-Binding Cassette Transporters ,Female ,T-Lymphocytes, Cytotoxic - Abstract
The absence of tumor-associated antigens (TAA) which might elicit an immune response is one reason for the disappointing results of therapeutical vaccines in cancer patients. Moreover, impaired expression of MHC class-I and components involved in antigen processing, such as TAP-1, -2, LMP-2, -7, and MECL-1, may lead to tumor escape from immune recognition. Expression profiling of TAA is one approach towards the design of well-defined and individualized anti-tumor vaccines. Quantitative polymerase chain reaction (qRT-PCR) is the method of choice to characterize immunologically relevant properties of individual tumors. However, the application of qRT-PCR as a surrogate parameter for the expression of TAAs depends upon the assumption that the level of an mRNA species correlates with the cellular level of the protein it encodes. Therefore, we additionally analyzed TAA expression by immunofluorescence and T cell recognition. In the present study we were unable to confirm that impaired TAP-1 or -2 (transporter associated with antigen processing) expression characterized at the mRNA level is an appropriate surrogate parameter for down-regulated MHC class-I expression in breast cancer. In addition, we analyzed the expression pattern of TAAs in breast and ovarian cancer cell lines. Besides the well-known over-expression of HER-2/neu, CEA, and MUC-1, multiple antigens of the MAGE-family were frequently co-expressed. We investigated whether detection of TAAs by qRT-PCR correlates with monoclonal antibody staining, and which method could predict T cell recognition. We demonstrated a correlation between tumor cell lysis by HLA-A*0201-restricted, MUC-1-specific CTL and threshold levels of MUC-1-specific mRNA. MUC-1 is an example that TAA profiling by RT-PCR and flow cytometry can fail to correlate with each other and are of limited value in the prediction of T cell recognition.
- Published
- 2003
49. An essential role for tripeptidyl peptidase in the generation of an MHC class I epitope
- Author
-
Ulrike Seifert, Concepción Marañón, Ayelet Shmueli, Jean-François Desoutter, Lisa Wesoloski, Katharina Janek, Peter Henklein, Susanne Diescher, Muriel Andrieu, Henri de la Salle, Toni Weinschenk, Hansjörg Schild, Diego Laderach, Anne Galy, Gaby Haas, Peter-M. Kloetzel, Yuval Reiss, and Anne Hosmalin
- Subjects
Proteasome Endopeptidase Complex ,Immunology ,Antigen presentation ,Major histocompatibility complex ,Aminopeptidases ,Tripeptidyl peptidase ,Epitope ,Epitopes ,Multienzyme Complexes ,Endopeptidases ,MHC class I ,Humans ,Immunology and Allergy ,RNA, Small Interfering ,Dipeptidyl-Peptidases and Tripeptidyl-Peptidases ,Antigen Presentation ,biology ,Antigen processing ,Histocompatibility Antigens Class I ,HIV ,Tripeptidyl peptidase II ,Dendritic Cells ,Virology ,Anti-Bacterial Agents ,Cell biology ,Cysteine Endopeptidases ,Proteasome ,biology.protein ,Oligopeptides - Abstract
Most of the peptides presented by major histocompatibility complex (MHC) class I molecules require processing by proteasomes. Tripeptidyl peptidase II (TPPII), an aminopeptidase with endoproteolytic activity, may also have a role in antigen processing. Here, we analyzed the processing and presentation of the immunodominant human immunodeficiency virus epitope HIV-Nef(73-82) in human dendritic cells. We found that inhibition of proteasome activity did not impair Nef(73-82) epitope presentation. In contrast, specific inhibition of TPPII led to a reduction of Nef(73-82) epitope presentation. We propose that TPPII can act in combination with or independent of the proteasome system and can generate epitopes that evade generation by the proteasome-system.
- Published
- 2003
50. Release of regulators of angiogenesis following Hypocrellin-A and -B photodynamic therapy of human brain tumor cells
- Author
-
Toni Weinschenk, Martin H. Deininger, Hermann J. Schluesener, Richard Meyermann, and Matthias Morgalla
- Subjects
Angiogenesis ,Blotting, Western ,Biophysics ,Biology ,Vascular endothelial growth inhibitor ,Biochemistry ,Mass Spectrometry ,chemistry.chemical_compound ,Tumor Cells, Cultured ,Humans ,education ,Perylene ,Molecular Biology ,education.field_of_study ,Photosensitizing Agents ,Phenol ,Brain Neoplasms ,Quinones ,Cell Biology ,Vascular endothelial growth factor B ,Endothelial stem cell ,Vascular endothelial growth factor ,Vascular endothelial growth factor A ,Photochemotherapy ,Vascular endothelial growth factor C ,chemistry ,Immunology ,Allograft inflammatory factor 1 ,Cancer research ,Angiogenesis Inducing Agents ,Electrophoresis, Polyacrylamide Gel - Abstract
Photodynamic therapy (PDT) is an innovative strategy for the treatment of solid neoplasms of the brain. Aside from inducing cell death in tumor cells, PDT induces endothelial cell death and promotes formation of blood clots; however, exact mechanisms that trigger these phenomena remain largely unknown. We now used Western blotting to analyze secretion of regulators of angiogenesis to the supernatants of one glioma, one macrophage, and one endothelial cell line following Hypocrellin-A and -B photodynamic therapy. We observed induction of proangiogenic VEGF (vascular endothelial growth factor) and of antiangiogenic sFlt-1, angiostatin, p43, allograft inflammatory factor-1, and connective tissue growth factor. Release of thrombospondin-1 was diminished in a glioma cell line supernatant. Endostatin release was induced in glioma cells and reduced in macrophages and endothelial cells. These data show that a wide range of antiangiogenic factors are secreted by brain tumor cells following Hypocrellin photochemotherapy. However, VEGF release is also induced thus suggesting both favorable and deleterious effects on tumor outgrowth.
- Published
- 2002
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.