54 results on '"Harpreet Singh-Jasuja"'
Search Results
2. 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
3. Additional Tables and Figures from A Cancer Research UK First Time in Human Phase I Trial of IMA950 (Novel Multipeptide Therapeutic Vaccine) in Patients with Newly Diagnosed Glioblastoma
- Author
-
James W.A. Ritchie, Sarah E. Halford, Harpreet Singh-Jasuja, Oliver Schoor, Karen Hill, Jane Peters, Lesley McGuigan, Norbert Hilf, Sarah Kutscher, Juha Lindner, Willie Stewart, Alan Jackson, Sarah Jefferies, Catherine McBain, Christopher J. Twelves, Omar Al-Salihi, Allan James, Paul J. Mulholland, Sharon Peoples, and Roy Rampling
- Abstract
Supplementary Table S1. TUMAPs contained in IMA950 and associated source antigens. Supplementary Table S2. Statistical analysis underlying the recruitment of 20 immune evaluable patients per cohort. Supplementary Table S3. Reasons for HLA-A*02 positive patients not entering the clinical trial. Supplementary Figure S1. Study schedule. Supplementary Figure S2. Exemplary gating strategy used for the primary multimer assay. Supplementary Figure S3. Exemplary immune responses in patients as determined by multimer assay after in vitro sensitization. Supplementary Figure S4. Apparent diffusion coefficient (ADC) in Cohort 1 at each of the four scan points.
- Published
- 2023
4. 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
5. 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
6. Data from A Cancer Research UK First Time in Human Phase I Trial of IMA950 (Novel Multipeptide Therapeutic Vaccine) in Patients with Newly Diagnosed Glioblastoma
- Author
-
James W.A. Ritchie, Sarah E. Halford, Harpreet Singh-Jasuja, Oliver Schoor, Karen Hill, Jane Peters, Lesley McGuigan, Norbert Hilf, Sarah Kutscher, Juha Lindner, Willie Stewart, Alan Jackson, Sarah Jefferies, Catherine McBain, Christopher J. Twelves, Omar Al-Salihi, Allan James, Paul J. Mulholland, Sharon Peoples, and Roy Rampling
- Abstract
Purpose: To perform a two-cohort, phase I safety and immunogenicity study of IMA950 in addition to standard chemoradiotherapy and adjuvant temozolomide in patients with newly diagnosed glioblastoma. IMA950 is a novel glioblastoma-specific therapeutic vaccine containing 11 tumor-associated peptides (TUMAP), identified on human leukocyte antigen (HLA) surface receptors in primary human glioblastoma tissue.Experimental Design: Patients were HLA-A*02–positive and had undergone tumor resection. Vaccination comprised 11 intradermal injections with IMA950 plus granulocyte macrophage colony-stimulating factor (GM-CSF) over a 24-week period, beginning 7 to 14 days prior to initiation of chemoradiotherapy (Cohort 1) or 7 days after chemoradiotherapy (Cohort 2). Safety was assessed according to NCI CTCAE Version 4.0 and TUMAP-specific T-cell immune responses determined. Secondary observations included progression-free survival (PFS), pretreatment regulatory T cell (Treg) levels, and the effect of steroids on T-cell responses.Results: Forty-five patients were recruited. Related adverse events included minor injection site reactions, rash, pruritus, fatigue, neutropenia and single cases of allergic reaction, anemia and anaphylaxis. Two patients experienced grade 3 dose-limiting toxicity of fatigue and anaphylaxis. Of 40 evaluable patients, 36 were TUMAP responders and 20 were multi-TUMAP responders, with no important differences between cohorts. No effect of pretreatment Treg levels on IMA950 immunogenicity was observed, and steroids did not affect TUMAP responses. PFS rates were 74% at 6 months and 31% at 9 months.Conclusions: IMA950 plus GM-CSF was well-tolerated with the primary immunogenicity endpoint of observing multi-TUMAP responses in at least 30% of patients exceeded. Further development of IMA950 is encouraged. Clin Cancer Res; 22(19); 4776–85. ©2016 AACR.See related commentary by Lowenstein and Castro, p. 4760
- Published
- 2023
7. 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
8. 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
9. 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
10. A Cancer Research UK First Time in Human Phase I Trial of IMA950 (Novel Multipeptide Therapeutic Vaccine) in Patients with Newly Diagnosed Glioblastoma
- Author
-
Paul Mulholland, Oliver Schoor, Omar Al-Salihi, Chris Twelves, Juha Lindner, Harpreet Singh-Jasuja, Sharon Peoples, Alan Jackson, Sarah Halford, James W A Ritchie, Lesley McGuigan, Catherine McBain, Roy Rampling, Sarah Kutscher, Allan James, Willie Stewart, Norbert Hilf, Jane Peters, Sarah Jefferies, and Karen Hill
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,T-Lymphocytes ,medicine.medical_treatment ,Antineoplastic Agents ,Kaplan-Meier Estimate ,Neutropenia ,Lymphocyte Activation ,Cancer Vaccines ,Disease-Free Survival ,Article ,Epitopes ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Antigens, Neoplasm ,Internal medicine ,medicine ,Humans ,Adverse effect ,Aged ,Temozolomide ,Brain Neoplasms ,business.industry ,Immunogenicity ,Glioma ,Chemoradiotherapy ,biochemical phenomena, metabolism, and nutrition ,Middle Aged ,medicine.disease ,Rash ,United Kingdom ,Clinical trial ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cohort ,Immunology ,Female ,medicine.symptom ,Glioblastoma ,Peptides ,business ,Adjuvant ,medicine.drug - Abstract
Purpose: To perform a two-cohort, phase I safety and immunogenicity study of IMA950 in addition to standard chemoradiotherapy and adjuvant temozolomide in patients with newly diagnosed glioblastoma. IMA950 is a novel glioblastoma-specific therapeutic vaccine containing 11 tumor-associated peptides (TUMAP), identified on human leukocyte antigen (HLA) surface receptors in primary human glioblastoma tissue. Experimental Design: Patients were HLA-A*02–positive and had undergone tumor resection. Vaccination comprised 11 intradermal injections with IMA950 plus granulocyte macrophage colony-stimulating factor (GM-CSF) over a 24-week period, beginning 7 to 14 days prior to initiation of chemoradiotherapy (Cohort 1) or 7 days after chemoradiotherapy (Cohort 2). Safety was assessed according to NCI CTCAE Version 4.0 and TUMAP-specific T-cell immune responses determined. Secondary observations included progression-free survival (PFS), pretreatment regulatory T cell (Treg) levels, and the effect of steroids on T-cell responses. Results: Forty-five patients were recruited. Related adverse events included minor injection site reactions, rash, pruritus, fatigue, neutropenia and single cases of allergic reaction, anemia and anaphylaxis. Two patients experienced grade 3 dose-limiting toxicity of fatigue and anaphylaxis. Of 40 evaluable patients, 36 were TUMAP responders and 20 were multi-TUMAP responders, with no important differences between cohorts. No effect of pretreatment Treg levels on IMA950 immunogenicity was observed, and steroids did not affect TUMAP responses. PFS rates were 74% at 6 months and 31% at 9 months. Conclusions: IMA950 plus GM-CSF was well-tolerated with the primary immunogenicity endpoint of observing multi-TUMAP responses in at least 30% of patients exceeded. Further development of IMA950 is encouraged. Clin Cancer Res; 22(19); 4776–85. ©2016 AACR. See related commentary by Lowenstein and Castro, p. 4760
- Published
- 2016
11. 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
12. The European regulatory environment of rna-based vaccines
- Author
-
Kajo Kallen, Hans-Georg Rammensee, Ulrich Kalinke, Thomas Hinz, Christoph Huber, Cedrik M. Britten, Sebastian Kreiter, Samir N. Khleif, Ulrich Granzer, Axel Hoos, Bruno Flamion, Özlem Türeci, Harpreet Singh-Jasuja, and Ugur Sahin
- Subjects
0301 basic medicine ,Autologous cell ,Messenger RNA ,Vaccines ,Anticancer vaccination ,Genetically modified medicinal products ,business.industry ,Genetic enhancement ,mRNA ,RNA ,Genetic therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Antigen ,Preventive and therapeutic approaches ,Infectious disease (medical specialty) ,030220 oncology & carcinogenesis ,Advanced therapy medicinal products (ATMP) ,Immunology ,Medicine ,Vaccination against infectious disease ,business ,Regulatory framework in the EU ,Ex vivo - Abstract
A variety of different mRNA-based drugs are currently in development. This became possible, since major breakthroughs in RNA research during the last decades allowed impressive improvements of translation, stability and delivery of mRNA. This article focuses on antigen-encoding RNA-based vaccines that are either directed against tumors or pathogens. mRNA-encoded vaccines are developed both for preventive or therapeutic purposes. Most mRNA-based vaccines are directly administered to patients. Alternatively, primary autologous cells from cancer patients are modified ex vivo by the use of mRNA and then are adoptively transferred to patients. In the EU no regulatory guidelines presently exist that specifically address mRNA-based vaccines. The existing regulatory framework, however, clearly defines that mRNA-based vaccines in most cases have to be centrally approved. Interestingly, depending on whether RNA-based vaccines are directed against tumors or infectious disease, they are formally considered gene therapy products or not, respectively. Besides an overview on the current clinical use of mRNA vaccines in various therapeutic areas a detailed discussion of the current regulatory situation is provided and regulatory perspectives are discussed.
- Published
- 2017
13. The European Regulatory Environment of RNA-Based Vaccines
- Author
-
Thomas, Hinz, Kajo, Kallen, Cedrik M, Britten, Bruno, Flamion, Ulrich, Granzer, Axel, Hoos, Christoph, Huber, Samir, Khleif, Sebastian, Kreiter, Hans-Georg, Rammensee, Ugur, Sahin, Harpreet, Singh-Jasuja, Özlem, Türeci, and Ulrich, Kalinke
- Subjects
Europe ,Neoplasms ,Animals ,Humans ,Genetic Therapy ,RNA, Messenger ,Antigens ,Cancer Vaccines - Abstract
A variety of different mRNA-based drugs are currently in development. This became possible, since major breakthroughs in RNA research during the last decades allowed impressive improvements of translation, stability and delivery of mRNA. This article focuses on antigen-encoding RNA-based vaccines that are either directed against tumors or pathogens. mRNA-encoded vaccines are developed both for preventive or therapeutic purposes. Most mRNA-based vaccines are directly administered to patients. Alternatively, primary autologous cells from cancer patients are modified ex vivo by the use of mRNA and then are adoptively transferred to patients. In the EU no regulatory guidelines presently exist that specifically address mRNA-based vaccines. The existing regulatory framework, however, clearly defines that mRNA-based vaccines in most cases have to be centrally approved. Interestingly, depending on whether RNA-based vaccines are directed against tumors or infectious disease, they are formally considered gene therapy products or not, respectively. Besides an overview on the current clinical use of mRNA vaccines in various therapeutic areas a detailed discussion of the current regulatory situation is provided and regulatory perspectives are discussed.
- Published
- 2016
14. 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
15. Corrigendum to 'New vaccination strategies in liver cancer' [Cytokine Growth Factor Rev. 36 (2017) 125–129]
- Author
-
Luigi Buonaguro, Andrea Mayer-Mokler, Christian Flohr, Carsten Reinhardt, Harpreet Singh-Jasuja, Roberto Accolla, Giovanna Tosi, Yuk T. Ma, David Adams, Danila Valmori, Regina Heidenreich, Ulrike Gnad-Vogt, Alfred Königsrainer, Markus Löffler, Hans-Georg Rammensee, Bruno Sangro, Sven Francque, Maria Tagliamonte, Annacarmen Petrizzo, Maria Lina Tornesello, and Franco M. Buonaguro
- Subjects
business.industry ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Growth factor ,Immunology ,medicine.disease ,Virology ,General Biochemistry, Genetics and Molecular Biology ,Vaccination ,Cytokine ,Immunology and Allergy ,Medicine ,business ,Liver cancer - Published
- 2019
16. HLA ligandome tumor antigen discovery for personalized vaccine approach
- Author
-
Harpreet Singh-Jasuja and Hans-Georg Rammensee
- Subjects
medicine.medical_treatment ,Immunology ,T cells ,Human leukocyte antigen ,Review ,Biology ,immunogenicity ,medicine.disease_cause ,Cancer Vaccines ,Cancer immunotherapy ,Antigen ,antigens ,immunomonitoring ,Antigens, Neoplasm ,Neoplasms ,Drug Discovery ,medicine ,Humans ,Precision Medicine ,Pharmacology ,Mutation ,cancer immunotherapy ,Cancer ,Immunotherapy ,medicine.disease ,mutations ,vaccination ,Phosphoproteins ,Tumor antigen ,genome sequencing ,Cancer cell ,Molecular Medicine ,HLA ligandome - Abstract
Every cancer is different and cancer cells differ from normal cells, in particular, through genetic alterations. HLA molecules on the cell surface enable T lymphocytes to recognize cellular alterations as antigens, including mutations, increase in gene product copy numbers or expression of genes usually not used in the adult organism. The search for cancer-associated antigens shared by many patients with a particular cancer has yielded a number of hits used in clinical vaccination trials with indication of survival benefit. Targeting cancer-specific antigens, which are exclusively expressed on cancer cells and not on normal cells, holds the promise for much better results and perhaps even a cure. Such antigens, however, may specifically appear in very few patients or may be mutated appearing just in one patient. Therefore, to target these in a molecularly defined way, the approach has to be individualized.
- Published
- 2013
17. Targeted therapy in renal cell carcinoma: moving from molecular agents to specific immunotherapy
- Author
-
Jens Bedke, Harpreet Singh-Jasuja, Cécile Gouttefangeas, Stefan Stevanovic, C.L. Behnes, and Arnulf Stenzl
- Subjects
Vascular Endothelial Growth Factor A ,IMA901 ,Cyclophosphamide ,medicine.drug_class ,Urology ,medicine.medical_treatment ,Tyrosine kinase inhibitor ,Antineoplastic Agents ,Cancer Vaccines ,Tyrosine-kinase inhibitor ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Renal cell carcinoma ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Molecular Targeted Therapy ,Carcinoma, Renal Cell ,030304 developmental biology ,0303 health sciences ,business.industry ,Sunitinib ,TOR Serine-Threonine Kinases ,Vaccination ,Immunotherapy ,Topic Paper ,medicine.disease ,Kidney Neoplasms ,Immune checkpoint ,3. Good health ,Cytokine ,Immune therapy ,030220 oncology & carcinogenesis ,Cancer research ,business ,medicine.drug - Abstract
Non-specific immunotherapy has been for a long time a standard treatment option for patients with metastatic renal cell carcinoma but was redeemed by specific targeted molecular therapies, namely the VEGF and mTOR inhibitors. After moving treatment for mRCC to specific molecular agents with a well-defined mode of action, immunotherapy still needs this further development to increase its accuracy. Nowadays, an evolution from a rather non-specific cytokine treatment to sophisticated targeted approaches in specific immunotherapy led to a re-launch of immunotherapy in clinical studies. Recent steps in the development of immunotherapy strategies are discussed in this review with a special focus on peptide vaccination which aims at a tumor targeting by specific T lymphocytes. In addition, different combinatory strategies with immunomodulating agents like cyclophosphamide or sunitinib are outlined, and the effects of immune checkpoint modulators as anti-CTLA-4 or PD-1 antibodies are discussed. peerReviewed
- Published
- 2013
18. 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
19. ATIM-20. GAPVAC-101 TRIAL OF A HIGHLY PERSONALIZED PEPTIDE VACCINATION FOR PATIENTS WITH NEWLY DIAGNOSED GLIOBLASTOMA
- Author
-
Andreas von Deimling, Hideho Okada, Wolfgang Wick, Cécile Gouttefangeas, Michael Platten, Harpreet Singh-Jasuja, Katrin Frenzel, Sjoerd H. van der Burg, Christian H. Ottensmeier, Arie Admon, Pierre-Yves Dietrich, Judith R. Kroep, Berta Ponsati, Ugur Sahin, Hans-Georg Rammensee, Hans Skovgaard Poulsen, Francisco Martínez-Ricarte, Ghazaleh Tabatabai, Stefan Stevanovic, and Norbert Hilf
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,Standard of care ,business.industry ,medicine.medical_treatment ,Cancer ,Newly diagnosed ,Immunotherapy ,medicine.disease ,Vaccination ,Abstracts ,Internal medicine ,medicine ,Neurology (clinical) ,Adverse effect ,business ,medicine.drug ,Glioblastoma - Abstract
BACKGROUND: There is a need for treatment personalization as every cancer is molecularly unique. In addition glioblastoma (GB) are immunologically regarded as resistant, cold tumor with few targetable antigens available from mutations, thus demanding new personalized immunotherapies. So far outside Neuro-Oncology, T cells orchestrate impressive anti-tumor effects with checkpoint inhibitors, but also vaccines. METHODS: The GAPVAC consortium established an immunotherapy, for which personalized selection of 2 peptide-based actively personalized vaccines (APVAC) per patient for treatment of newly diagnosed GB was based not only on whole-exome sequencing but also on human leukocyte antigen (HLA)-ligandome analyses providing insight into the actual presentation of relevant epitopes in the tumor. GAPVAC-101 (NCT02149225) enrolled 16 patients in a European phase I feasibility, safety and immunogenicity trial integrated into standard of care. For APVAC1, up to 7 peptides were selected from a trial specific warehouse based on individual biomarker data. Vaccination (i.d.) with GM-CSF and poly-ICLC in 15 patients started with the 1st adjuvant cycle of temozolomide (TMZ). For APVAC2, analyses revealed a median of 36 somatic, non-synonymous mutations in the patients tumors. From the 4th TMZ cycle, 11 patients received APVAC2 with usually 2 de novo antigens per patient selected according to mutation, actual or putative HLA presentation and immunogenicity. Overall 20 APVAC2 antigens incl. 14 mutated were vaccinated. RESULTS: Adverse events were largely reversible injection site reactions and two anaphylactic reactions and one increase in cerebral edema. Short, non-mutated APVAC1 antigens induced sustained CD8 responses with memory phenotype. Mutated APVAC2 antigens induced predominantly CD4 responses of favorable TH1 type. Median PFS and OS were 14.2 and 29 months from diagnosis, respectively, in patients that received 1 APVAC vaccination (N=15). CONCLUSION: Overall, GAPVAC displayed expected safety profiles and high biological activity indicating further development.
- Published
- 2018
20. OS2.2 Highly personalized peptide vaccination for patients with newly diagnosed glioblastoma: the GAPVAC trial
- Author
-
Cécile Gouttefangeas, Christian H. Ottensmeier, Hans Skovgaard Poulsen, Norbert Hilf, Valesca Bukur, Ulrik Lassen, Berta Ponsati, Judith R. Kroep, P thor Straten, Jordi Rodon, Michael Platten, Ugur Sahin, H Okada, Ghazaleh Tabatabai, Pierre-Yves Dietrich, Stefan Stevanovic, Arie Admon, Wolfgang Wick, Harpreet Singh-Jasuja, A. von Deimling, Francisco Martínez-Ricarte, H.-G. Rammensee, Katrin Frenzel, and S.H. van der Burg
- Subjects
Oncology ,chemistry.chemical_classification ,Cancer Research ,medicine.medical_specialty ,business.industry ,Peptide ,Newly diagnosed ,medicine.disease ,Vaccination ,chemistry ,Internal medicine ,Oral Presentations ,medicine ,Neurology (clinical) ,business ,Glioblastoma - Abstract
BACKGROUND: There is a need for treatment personalization as every cancer is molecularly unique. In addition glioblastoma (GB) are immunologically regarded as resistant, “cold” tumor with few targetable antigens available from mutations, thus demanding new personalized immunotherapies. So far outside Neuro-Oncology, T cells orchestrate impressive anti-tumor effects with checkpoint inhibitors, but also vaccines. MATERIAL AND METHODS: The GAPVAC consortium established an immunotherapy, for which personalized selection of 2 peptide-based actively personalized vaccines (APVAC) per patient for treatment of newly diagnosed GB was based not only on whole-exome sequencing but also on human leukocyte antigen (HLA)-ligandome analyses providing insight into the actual presentation of relevant epitopes in the tumor. GAPVAC-101 (NCT02149225) enrolled 16 patients in a European phase I feasibility, safety and immunogenicity trial integrated into standard of care. For APVAC1, up to 7 peptides were selected from a trial specific warehouse based on individual biomarker data. Vaccination (i.d.) with GM-CSF and poly-ICLC in 15 patients started with the 1(st) adjuvant cycle of temozolomide (TMZ). For APVAC2, analyses revealed a median of 36 somatic, non-synonymous mutations in the patients’ tumors. From the 4(th) TMZ cycle, 11 patients received APVAC2 with usually 2 de novo antigens per patient selected according to mutation, actual or putative HLA presentation and immunogenicity. Overall 20 APVAC2 antigens incl. 14 mutated were vaccinated. RESULTS: Adverse events were largely reversible injection site reactions and two anaphylactic reactions and one increase in cerebral edema. Short, non-mutated APVAC1 antigens induced sustained CD8 responses with memory phenotype. Mutated APVAC2 antigens induced predominantly CD4 responses of favorable TH1 type. Median PFS and OS were 14.2 and 29 months from diagnosis, respectively, in patients that received ≥1 APVAC vaccination (N=15). CONCLUSION: Overall, GAPVAC displayed expected safety profiles and high biological activity indicating further development.
- Published
- 2018
21. 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
22. 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
-
Norbert Hilf, Barbara Rakitsch, Helen Hörzer, Arun Satelli, Franziska Hoffgaard, Oliver Schoor, Harpreet Singh-Jasuja, Weinschenk Toni, and Jens Fritsche
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Human leucocyte antigen ,business.industry ,medicine.medical_treatment ,Cancer ,Target peptide ,Human leukocyte antigen ,medicine.disease ,Cancer immunotherapy ,Internal medicine ,Gene expression ,medicine ,Adoptive cellular therapy ,business ,Companion diagnostic - Abstract
Adoptive cellular therapy (ACT) has dramatically changed the landscape of cancer immunotherapy. ACTolog® and ACTengine® are actively personalized ACT approaches employing T-cell receptor products based on a warehouse of human leucocyte antigen (HLA)-bound peptide targets. Selecting the relevant target candidates requires the establishment of biomarkers predictive for HLA peptide presentation and their development into companion diagnostic devices. Here we describe the development of IMA_Detect, a diagnostic test based on gene expression analysis of primary patient tumor material by qPCR which is predictive for presentation of a target peptide by HLA. To establish mRNA expression levels which indicate actual peptide presentation, data of Immatics' XPRESIDENT® target discovery platform was used by integrating quantitative immunopeptidomics data (label-free LC-MS) with paired transcriptomics data (RNA-Seq). The peptide-specific correlation between peptide presentation and expression of the coding exons was verified for each target individually. The resulting RNA-Seq thresholds were mapped to qPCR thresholds using calibration curves followed by validation of the qPCR assay. The IMA_Detect test is performed in a CLIA/CAP approved setting and is currently being applied in the phase I ACT trials IMA101-101 and IMA201-101 to determine whether a patient's tumor expresses any of the tested targets at levels considered sufficient for potential benefit from the administered T-cell therapy. IMA101-101 uses autologous T-cell products (ACTolog®) for patients with solid cancers while IMA201-101 is based on TCR-engineered T Cells (ACTengine®) in NSCLC and HNSCC patients. We will present first results of the patient screening and the personalized target selection. Citation Format: Jens Fritsche, Arun Satelli, Helen Hörzer, Barbara Rakitsch, Franziska Hoffgaard, Norbert Hilf, Oliver Schoor, Harpreet Singh-Jasuja, Weinschenk Toni. IMA_Detect: Mass spectrometry guided development and clinical application of a companion diagnostic for adoptive cellular therapy against tumor associated HLA peptides [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 662.
- Published
- 2018
23. 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
24. MP16-11 JAPANESE PHASE I/II STUDY OF MULTIPEPTIDE-BASED CANCER VACCINE IMA901 AFTER SINGLE-DOSE CYCLOPHOSPHAMIDE IN JAPANESE PATIENTS WITH ADVANCED RENAL CELL CANCER
- Author
-
Hongo, Fumiya, primary, Takaha, Natsuki, additional, Ueda, Takashi, additional, Yano, Kimihiro, additional, Tamada, Satoshi, additional, Oliver, Schoor, additional, Harpreet, Singh-Jasuja, additional, Nakatani, Tatsuya, additional, Miki, Tsuneharu, additional, and Ukimura, Osamu, additional
- Published
- 2017
- Full Text
- View/download PDF
25. The regulatory landscape for actively personalized cancer immunotherapies
- Author
-
Cedrik M. Britten, Hans-Georg Rammensee, Harpreet Singh-Jasuja, Karl Josef Kallen, Ugur Sahin, Ulrich Kalinke, Christoph Huber, Samir N. Khleif, Axel Hoos, Michaela Nielsen, Sebastian Kreiter, Bruno Flamion, and Thomas Hinz
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Biomedical Engineering ,Cancer ,Bioengineering ,medicine.disease ,Cancer Vaccines ,Applied Microbiology and Biotechnology ,Social Control, Formal ,Drug development ,Neoplasms ,Internal medicine ,Immunology ,Biomarkers, Tumor ,medicine ,Humans ,Molecular Medicine ,Immunotherapy ,Precision Medicine ,business ,Biotechnology - Published
- 2013
26. Selective intracellular retention of virally induced NKG2D ligands by the human cytomegalovirus UL16 glycoprotein
- Author
-
Stefan Z. Lutz, Harpreet Singh-Jasuja, Alexander Steinle, Kerstin Laib Sampaio, Stefan Welte, Christian Sinzger, Ute Eknigk, and Hans-Georg Rammensee
- Subjects
Human cytomegalovirus ,NK Cell Lectin-Like Receptor Subfamily K ,viruses ,Immunology ,Cytomegalovirus ,Down-Regulation ,chemical and pharmacologic phenomena ,Biology ,GPI-Linked Proteins ,Transfection ,Cell Line ,Natural killer cell ,Viral Proteins ,medicine ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Receptors, Immunologic ,Endoplasmic reticulum ,Histocompatibility Antigens Class I ,Intracellular Signaling Peptides and Proteins ,Membrane Proteins ,Fibroblasts ,Flow Cytometry ,NKG2D ,medicine.disease ,Molecular biology ,Cell biology ,Killer Cells, Natural ,Protein Transport ,ULBP1 ,ULBP2 ,medicine.anatomical_structure ,Gene Expression Regulation ,Cytomegalovirus Infections ,Intercellular Signaling Peptides and Proteins ,Receptors, Natural Killer Cell ,Carrier Proteins - Abstract
Human cytomegalovirus (HCMV) has evolved a multitude of molecular mechanisms to evade the antiviral immune defense of the host. Recently, using soluble recombinant molecules, the HCMV UL16 glycoprotein was shown to interact with some ligands of the activating immunoreceptor NKG2D and, therefore, may also function as a viral immunomodulator. However, the role of UL16 during the course of HCMV infection remained unclear. Here, we demonstrate that HCMV infection of fibroblasts induces expression of all known NKG2D ligands (NKG2DL). However, solely MICA and ULBP3 reach the cellular surface to engage NKG2D, whereas MICB, ULBP1 and ULBP2 are selectively retained in the endoplasmic reticulum by UL16. UL16-mediated reduction of NKG2DL cell surface density diminished NK cytotoxicity. Thus, UL16 functions by capturing activating ligands for cytotoxic lymphocytes that are synthesized in response to HCMV infection.
- Published
- 2003
27. The heat shock protein Gp96 links innate and specific immunity
- Author
-
Norbert Hilf, Harpreet Singh-Jasuja, and Hansjörg Schild
- Subjects
Cancer Research ,Fever ,Physiology ,T-Lymphocytes ,Antigen presentation ,chemical and pharmacologic phenomena ,Dendritic Cells ,Biology ,Acquired immune system ,Immune system ,Antigen ,Antigens, Neoplasm ,Immune System ,Physiology (medical) ,Heat shock protein ,Immunology ,MHC class I ,biology.protein ,Animals ,Cytotoxic T cell ,Antigen-presenting cell - Abstract
Among other heat shock proteins (HSPs), the ER-resident chaperone Gp96 has been described as a potent tumour vaccine in animal models. A growing list of data underlines that Gp96 triggers both arms of pathogen defence-innate and specific immunity-in a synergistic and most efficient way: It enables specific immune responses by transferring immunogenic peptides that have been acquired in the ER to the MHC class I pathway of antigen presenting cells (APCs). For this, two important features of Gp96 are required. First, its ability to bind immunogenic peptides. Secondly, its acquisition by specialized antigen presenting cells capable of inducing cellular immune responses. Due to specific receptors on the surface of APCs, this uptake from the extracellular space occurs very efficiently and rapidly. Serving the innate branch of immunity, Gp96 unspecifically activates APCs, which then provide a pro-inflammatory cytokine milieu and co-stimulation to cytotoxic T cells. Thus, Gp96 uses all resources of the immune system to trigger cytotoxic T cell responses against associated peptides.
- Published
- 2002
28. Multi-peptide cancer vaccines for clinical application
- Author
-
Toni Weinschenk, Steffen Walter, and Harpreet Singh-Jasuja
- Subjects
chemistry.chemical_classification ,chemistry ,business.industry ,Cancer research ,medicine ,Cancer ,Peptide ,medicine.disease ,business - Published
- 2014
29. Evolution of the Regulatory Landscape for Immunomodulatory Compounds and Personalized Therapeutic Cancer Vaccines
- Author
-
Cedrik M. Britten, Ulrich Kalinke, Harpreet Singh-Jasuja, and Thomas Hinz
- Subjects
business.industry ,medicine.medical_treatment ,Cancer therapy ,Cancer ,Immunotherapy ,Biology ,medicine.disease ,Bioinformatics ,Biotechnology ,Cell therapy ,Safety profile ,Cancer immunotherapy ,medicine ,media_common.cataloged_instance ,Cancer vaccine ,European union ,business ,media_common - Abstract
The term cancer immunotherapy covers a huge range of different therapeutic interventions based on highly diverse medicinal products. Some immunotherapy products such as monoclonal antibodies have successfully been developed during the last years and contributed to major advances in cancer therapy. Development of cancer vaccines seemed to be more challenging. Nevertheless, some promising candidates are currently in late-stage clinical development, and one product (sipuleucel-T) was recently approved in the United States (USA) and in the European Union (EU). Additionally, cell therapy approaches based on adoptive lymphocyte transfer have recently come into focus due to surprisingly efficient antitumor effects on one side and a comparably good safety profile on the other. Here, we briefly summarize the regulatory classification of immunotherapy approaches. The second part focuses on the evolving regulatory landscape for nonpersonalized and personalized cancer vaccines that have been in the focus of the CIMT regulatory research group during the last years.
- Published
- 2014
30. Recent Developments in the Active Immunotherapy of Renal Cell Cancer
- Author
-
Harpreet Singh-Jasuja
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Angiogenesis ,medicine.medical_treatment ,Immunotherapy ,Active immunotherapy ,urologic and male genital diseases ,medicine.disease ,female genital diseases and pregnancy complications ,Cancer immunotherapy ,Renal cell carcinoma ,Internal medicine ,medicine ,Adenocarcinoma ,Cell cancer ,business ,neoplasms ,Kidney cancer - Abstract
Renal cell carcinoma (RCC) is the adenocarcinoma of the kidney and the most common form of kidney cancer. More than 300,000 people are newly affected by RCC every year globally (Ferlay et al. 2012). The most common subtype of RCC is clear-cell RCC which comprises about 75 % of RCCs. For two decades, cytokine-based immunotherapy was the standard of care for the management of RCC. In the last 10 years, a better understanding of the tumor biology of RCC has led to the approval of a number of novel agents targeting angiogenesis and has led to an improvement of patient outcomes. However, almost all patients eventually develop a resistance to antiangiogenic agents, and the extension of overall survival (by each individual agent on its own) has remained relatively modest. These observations as well as recent remarkable developments in the field of cancer immunotherapy have strongly resurged the interest in immunotherapeutic approaches for treatment of RCC. This chapter summarizes the recent developments in the treatment of RCC through immunotherapies approved or in clinical development as well as the immunological role of targeted therapies.
- Published
- 2014
31. The heat shock protein gp96 induces maturation of dendritic cells and down-regulation of its receptor
- Author
-
Harpreet Singh-Jasuja, Hans Scherer, Hans-Georg Rammensee, Norbert Hilf, Hansjörg Schild, Danièle Arnold-Schild, and René E. M. Toes
- Subjects
MHC class II ,T cell ,Immunology ,B-cell receptor ,Dendritic cell ,Biology ,Cell biology ,medicine.anatomical_structure ,medicine ,biology.protein ,Immunology and Allergy ,Cytokine secretion ,Scavenger receptor ,Receptor ,Mannose receptor - Abstract
Peptides associated with the heat shock protein gp96 induce a specific T cell response against cells from which gp96 is isolated. Recently, we have shown that gp96 binds to a yet unknown receptor present on dendritic cells (DC) and that receptor-mediated uptake is required for cross-presentation of gp96-associated peptides by DC. We now describe that gp96 mediates maturation of DC as determined by up-regulation of MHC class II and CD86 molecules, secretion of the cytokines IL-12 and TNF-alpha and enhanced T cell stimulatory capacity. Heat-denatured gp96 is not able to induce DC maturation and cytokine secretion. Furthermore, we show that mature DC are no longer able to bind gp96 molecules. Hence, the gp96 receptor is down-regulated on mature DC, suggesting that this receptor behaves similar to other receptors involved in antigen uptake like the scavenger receptor CD36, the mannose receptor or the integrins alpha(v)beta(3) and alpha(v)beta(5). Together, our findings provide an additional explanation for the remarkable immunogenicity of gp96 as a cross-priming antigen carrier and direct activator of DC.
- Published
- 2000
32. Cross-Presentation of Glycoprotein 96–Associated Antigens on Major Histocompatibility Complex Class I Molecules Requires Receptor-Mediated Endocytosis
- Author
-
Pieter Spee, Paola Ricciardi-Castagnoli, Jacques Neefjes, Christian Münz, René E. M. Toes, Hansjörg Schild, Stephen P. Schoenberger, Harpreet Singh-Jasuja, Norbert Hilf, Hans-Georg Rammensee, and Danièle Arnold-Schild
- Subjects
receptor-mediated endocytosis ,dendritic cell ,Immunology ,Antigen presentation ,Antigen-Presenting Cells ,cytotoxic T lymphocyte activation ,Receptors, Cell Surface ,Major histocompatibility complex ,Mice ,MHC class I ,Tumor Cells, Cultured ,Animals ,Humans ,Immunology and Allergy ,HSP70 Heat-Shock Proteins ,Adenovirus E1B Proteins ,Antigen-presenting cell ,Mice, Knockout ,Antigen Presentation ,B-Lymphocytes ,Mice, Inbred BALB C ,biology ,Macrophages ,Histocompatibility Antigens Class I ,H-2 Antigens ,Histocompatibility Antigens Class II ,Membrane Proteins ,Cross-presentation ,Dendritic Cells ,Receptor-mediated endocytosis ,Molecular biology ,Endocytosis ,Mice, Inbred C57BL ,CTL ,biology.protein ,Original Article ,cross-priming ,heat shock protein–peptide complex ,CD8 ,Molecular Chaperones - Abstract
Heat shock proteins (HSPs) like glycoprotein (gp)96 (glucose-regulated protein 94 [grp94]) are able to induce specific cytotoxic T lymphocyte (CTL) responses against cells from which they originate. Here, we demonstrate that for CTL activation by gp96-chaperoned peptides, specific receptor-mediated uptake of gp96 by antigen-presenting cells (APCs) is required. Moreover, we show that in both humans and mice, only professional APCs like dendritic cells (DCs), macrophages, and B cells, but not T cells, are able to bind gp96. The binding is saturable and can be inhibited using unlabeled gp96 molecules. Receptor binding by APCs leads to a rapid internalization of gp96, which colocalizes with endocytosed major histocompatibility complex (MHC) class I and class II molecules in endosomal compartments. Incubation of gp96 molecules isolated from cells expressing an adenovirus type 5 E1B epitope with the DC line D1 results in the activation of E1B-specific CTLs. This CTL activation can be specifically inhibited by the addition of irrelevant gp96 molecules not associated with E1B peptides. Our results demonstrate that only receptor-mediated endocytosis of gp96 molecules leads to MHC class I–restricted re-presentation of gp96-associated peptides and CTL activation; non–receptor-mediated, nonspecific endocytosis is not able to do so. Thus, we provide evidence on the mechanisms by which gp96 is participating in the cross-presentation of antigens from cellular origin.
- Published
- 1999
33. Single-dose cyclophosphamide synergizes with immune responses to the renal cell cancer vaccine IMA901
- Author
-
Toni Weinschenk, Carsten Reinhardt, Steffen Walter, and Harpreet Singh-Jasuja
- Subjects
Cyclophosphamide ,medicine.medical_treatment ,Immunology ,T cells ,renal cell cancer ,regulatory T cells ,Immune system ,immunomonitoring ,vaccine ,medicine ,Immunology and Allergy ,cancer ,Author's View ,business.industry ,Rationalization (psychology) ,biomarkers ,Immunotherapy ,randomized clinical trial ,Vaccination ,Oncology ,peptides ,low-dose cyclophosphamide ,Low dose cyclophosphamide ,Cell cancer ,immunotherapy ,business ,medicine.drug - Abstract
The development of efficient immunotherapies requires strong rationalization. We have recently implemented a large analysis of biomarkers in two studies involving the multi-peptide vaccine IMA901 and advanced renal cell cancer patients. Our findings demonstrate that the breadth of immune responses was associated with clinical benefits and that single-dose cyclophosphamide reduced the amount of regulatory T cells and was associated with prolonged survival after vaccination.
- Published
- 2013
34. Abstract 2291: On- and off target toxicity profiling for adoptive cell therapy by mass spectrometry-based immunopeptidome analysis of primary human normal tissues
- Author
-
Leonie Alten, Lea Stevermann, Toni Weinschenk, Jens Fritsche, Harpreet Singh-Jasuja, Hans-Georg Rammensee, Oliver Schoor, Annika Sonntag, Sarah Kutscher, Steffen Walter, Andrea Mahr, Valentina Goldfinger, Julia Leibold, Dominik Vahrenhorst, Franziska Hoffgaard, Dominik Maurer, and Sebastian Bunk
- Subjects
Cancer Research ,biology ,Colorectal cancer ,medicine.medical_treatment ,T-cell receptor ,Immunotherapy ,Human leukocyte antigen ,medicine.disease ,Epitope ,Cell therapy ,Carcinoembryonic antigen ,Oncology ,Immunology ,Proteome ,medicine ,biology.protein - Abstract
A major constraint for the broad and safe application of Adoptive Cellular Therapy (ACT) is the limited number of validated tumor targets, especially for solid tumors. For T-cell receptor (TCR)-based approaches, presentation of targeted HLA-peptides on normal tissues can lead to on-target toxicity, such as severe inflammatory colitis reported upon re-directing T cells to an HLA-A*02 restricted carcinoembryonic antigen (CEA) epitope. Independently, off-target cross-reactivity of TCRs occurred in previous ACT trials, e.g. when a MAGEA3-directed TCR cross-recognized an HLA-A*01 restricted epitope from titin expressed on heart, which led to fatal cardiac toxicities. Here we present a novel approach allowing the prediction of severe on- and off-target side effects before entering into clinical trials. We used a target discovery engine (XPRESIDENT) combining highly sensitive, quantitative mass spectrometry (LC-MS/MS), RNA-Seq-based differential transcriptomics, immunology and bioinformatics to characterize the human immunopeptidome directly on shock frozen primary human tissues. Over the last years we have built an according database for > 600 tumor samples from > 20 different tumor types and, importantly, > 300 samples from > 40 different normal tissue types, resulting in hundreds of thousands of unique HLA-peptide sequences. These data allow conclusions on which HLA peptides are actually presented on primary normal tissues in a quantitative manner, taking into account relative differences between normal tissues and tumors as well as absolute peptide copy numbers per cell. In order to assess the off-target risk for a TCR, we predict all theoretical HLA- and TCR-binding peptides in the proteome, ideally based on the binding motif of the TCR, and specifically search for actual peptide presentation by normal tissues. When analyzing the above described CEA case, we were able to detect the CEA-derived peptide IMIGVLVGV on HLA-A*02 positive colorectal cancer samples, but importantly also on normal colorectal samples. In the original study describing the titin case tremendous experimental efforts and sophisticated cell culture models were required to retrospectively identify cross-recognition of the peptide on cardiomyocytes as the cause of toxicity. In contrast, with our approach we easily and directly identified the critical peptide ESDPIVAQY as one of the most abundantly presented peptides on an HLA-A*01 positive primary human heart sample. We show that this approach can lead to noteworthy results also for other pre-clinical and clinical stage TCR candidates. In conclusion our data demonstrate that ultrasensitive LC-MS/MS of primary tissue may represent a fast, straightforward and meaningful complementary method to common in vitro or animal models for the prediction of on- and off-target toxicities in TCR-based immunotherapy approaches. Citation Format: Oliver Schoor, Jens Fritsche, Sarah Kutscher, Andrea Mahr, Lea Stevermann, Annika Sonntag, Franziska Hoffgaard, Dominik Vahrenhorst, Julia Leibold, Valentina Goldfinger, Leonie Alten, Sebastian Bunk, Dominik Maurer, Steffen Walter, Hans-Georg Rammensee, Harpreet Singh-Jasuja, Toni Weinschenk. On- and off target toxicity profiling for adoptive cell therapy by mass spectrometry-based immunopeptidome analysis of primary human normal tissues. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2291.
- Published
- 2016
35. Abstract 2354: Cancer vaccine development for hepatocellular carcinoma - HEPAVAC
- Author
-
Toni Weinschenk, Regina Heidenreich, Sven Francque, Sarah Kutscher, Bruno Sangro, Luigi Buonaguro, Alfred Koenigsrainer, Francesco Izzo, Harpreet Singh-Jasuja, Andrea Mayer, Markus W. Loeffler, Hans-Georg Rammensee, Roberto S. Accolla, Yuk Ting Ma, Phillip Mueller, and Danila Valmori
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Immunogenicity ,medicine.medical_treatment ,0206 medical engineering ,Cancer ,02 engineering and technology ,Human leukocyte antigen ,021001 nanoscience & nanotechnology ,medicine.disease ,020601 biomedical engineering ,Epitope ,Hepatocellular carcinoma ,Internal medicine ,CIITA ,medicine ,Cancer vaccine ,0210 nano-technology ,business ,Adjuvant - Abstract
The HEPAVAC Consortium aims to develop a highly innovative, novel cancer vaccine approach for hepatocellular carcinoma (HCC). The international project consortium consists of 9 European Partners from academia and the biotech industry with complementary and substantial expertise in developing immunotherapeutic strategies to treat cancer. The project has started in September 2013 and is supported by the European Commission's 7th Framework Program (www.hepavac.eu). HCC/normal adjacent tissue matched samples have been collected for HLA immunopeptidome analysis. 17 HCC samples from HLA-A*02+ patients and 15 samples from HLA-A*24+ patients have been analysed by mass spectrometry (LC-MS/MS). RNA-expression profiles have been established for 12 HCC samples. HLA-presentation/expression of peptides and mRNA on primary HCC samples are compared to >140 normal tissue samples from relevant organs (including heart, brain, lung, kidney, liver, nerve, skin etc.) from Immatics’ database. A total of 9051 HLA-A*02-restricted different tumor-associated peptides (TUMAPs) have been identified from HLA-A*02+ samples, while a total of 3286 different HLA-A*24-restricted TUMAPs have been identified from HLA-A*24+ samples. Of these, 33 HLA-A*02+ TUMAPS and 33 HLA-A*24+ TUMAPs have been validated, including peptide synthesis, immunogenicity testing and pharmaceutical evaluation. Most promising TUMAP candidates show selective expression only in HCC samples and no expression in normal tissues. In parallel, more than 6600 HLA-DR TUMAPs have been identified in Hep3B cells transfected with CIITA as well as an average of 1500 HLA-DR TUMAPs have been identified in HCC samples. A total of 16 newly identified HCC-specific epitopes have been selected for the HEPAVAC vaccine cocktail and are currently synthesized according to GMP standard. Of these, 7 are restricted to HLA- class I A*02; 5 HLA-A*24 and 4 HLA class II. In parallel, preclinical studies assessing the formulation and combination of the immunological RNA-based adjuvant (RNAdjuvant®) with peptide cocktails are underway. A multi Center phase I/II clinical trial in early HCC patients is predicted to start in July 2016. Citation Format: Sarah Kutscher, Roberto Accolla, Yuk T. Ma, Regina Heidenreich, Francesco Izzo, Alfred Koenigsrainer, Markus Loeffler, Phillip Mueller, Andrea Mayer, Hans-Georg Rammensee, Bruno Sangro, Sven Francque, Danila Valmori, Toni Weinschenk, Harpreet Singh-Jasuja, Luigi Buonaguro. Cancer vaccine development for hepatocellular carcinoma - HEPAVAC. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2354.
- Published
- 2016
36. Abstract 2654: GAPVAC-101 phase I trial: First data of an innovative actively personalized peptide vaccination trial in patients with newly diagnosed glioblastoma
- Author
-
Ghazaleh Tabatabai, Peter Lewandrowski, Monika Stieglbauer, Stefan Stevanovic, Sandra Heesch, Per thor Straten, Jordi Rodon, Ugur Sahin, Juan Sahuquillo, Colette Song, Valérie Dutoit, Randi Kristina Feist, Christian H. Ottensmeier, Francisco Martínez-Ricarte, Sabrina Kuttruff-Coqui, Hideho Okada, Bernhard Rössler, Judith R. Kroep, Wick Wolfgang, Jens Fritsche, Regina Mendryzk, Norbert Hilf, Karoline Laske, Hans-Georg Rammensee, Carsten Reinhardt, Marij Schoenmaekers-Welters, Harpreet Singh-Jasuja, Stevermann Lea, Toni Weinschenk, Michael Platten, Sjoerd H. van der Burg, Marco Skardelly, Katrin Frenzel, Ulrik Lassen, Christoph Huber, Valesca Bukur, Arie Admon, Berta Ponsati, David Capper, Hans Skovgaard Poulsen, Pierre-Yves Dietrich, Miriam Meyer, Anna Paruzynski, Nina Pawlowski, Sebastian Kreiter, Cécile Gouttefangeas, Andreas von Deimling, Martin Löwer, and Jordi Piro
- Subjects
0301 basic medicine ,Oncology ,Gerontology ,Cancer Research ,medicine.medical_specialty ,Phases of clinical research ,02 engineering and technology ,Newly diagnosed ,03 medical and health sciences ,Internal medicine ,medicine ,media_common.cataloged_instance ,In patient ,European union ,media_common ,Temozolomide ,business.industry ,021001 nanoscience & nanotechnology ,medicine.disease ,Gross Total Resection ,Vaccination ,030104 developmental biology ,0210 nano-technology ,business ,medicine.drug ,Glioblastoma - Abstract
The Glioma Actively Personalized Vaccine Consortium (GAPVAC; funded by the European Union Framework 7 Program) aims at treating newly diagnosed glioblastoma (GB) patients with two distinct actively personalized vaccines (APVACs). Resected tumor material is analyzed for multiple biomarkers to characterize the tumor in depth and to enable the design of APVACs tailored to each individual patient: Tumor-specific mutations, the HLA peptidome and gene expression profile are assessed by next-generation sequencing, mass spectrometry and RNA microarray analysis, respectively. Further, the patient-individual immune status is investigated by assessment of leukapheresis samples utilizing an in vitro immunogenicity platform. Data are integrated to define two distinct APVACs for each patient: APVAC1 is composed of up to ten peptides selected from a pre-manufactured “warehouse”. The warehouse contains 59 HLA class I-binding and three class II-binding tumor-associated peptides frequently over-presented in GB. APVAC2 is composed of one or two peptides that are de novo synthesized for a given patient and preferentially represent mutation-bearing neo-epitopes. After a preparation phase in which the warehouse was generated and setup of APVAC selection and manufacturing processes took place, the GAPVAC-101 phase I clinical trial was initiated. Primary endpoints of the study are assessment of safety, feasibility of APVAC manufacturing and biological activity. The trial is conducted at six European centers and recruits HLA-A*02:01 or A*24:02-positive patients with newly diagnosed GB after gross total resection. Patients receive APVAC1 and APVAC2 vaccinations plus immunomodulators (poly-ICLC and GM-CSF) three and six months post study enrolment, respectively, and concurrent to maintenance temozolomide (TMZ). As of November 2015, 11 patients have been enrolled, of whom six already received APVAC vaccines. Composition and manufacturing are ongoing for four patients. All APVACs were generated in time without ultimate failures. APVAC1 vaccines differ substantially with 31 out of 59 warehouse peptides have been selected at least once, indicating the need for personalization due to tumor heterogeneity even for non-mutated epitopes. In patients’ tumor samples an average of 40 non-synonymous mutations (including known driver mutations) were identified. Injection site reactions were the most frequent toxicities so far. One brain edema (Grade 3) and one allergic reaction (Grade 4)were observed, both potentially related to the vaccinations. First data on biological activity of APVACs and updated clinical data will be presented at the Annual Meeting. In conclusion, the GAPVAC concept has been successfully translated into the clinics and so far demonstrated to be safe and feasible with its level of personalization matching the observed tumor heterogeneity. Citation Format: Norbert Hilf, Katrin Frenzel, Sabrina Kuttruff-Coqui, Sandra Heesch, Sebastian Kreiter, Arie Admon, Valesca Bukur, Sjoerd van der Burg, Cecile Gouttefangeas, Judith R. Kroep, Marij Schoenmaekers-Welters, Jordi Piro, Berta Ponsati, Hans Skovgaard Poulsen, Ulrik Lassen, Francisco Martinez-Ricarte, Jordi Rodon, Juan Sahuquillo, Monika Stieglbauer, Stefan Stevanovic, Per thor Straten, Marco Skardelly, Ghazaleh Tabatabai, Michael Platten, David Capper, Andreas von Deimling, Valérie Dutoit, Hideho Okada, Christian Ottensmeier, Randi Kristina Feist, Jens Fritsche, Karoline Laske, Peter Lewandrowski, Martin Löwer, Regina Mendryzk, Miriam Meyer, Carsten Reinhardt, Bernhard Rössler, Anna Paruzynski, Nina Pawlowski, Colette Song, Stevermann Lea, Toni Weinschenk, Christoph Huber, Hans-Georg Rammensee, Pierre-Yves Dietrich, Wick Wolfgang, Ugur Sahin, Harpreet Singh-Jasuja. GAPVAC-101 phase I trial: First data of an innovative actively personalized peptide vaccination trial in patients with newly diagnosed glioblastoma. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2654.
- Published
- 2016
37. Abstract A115: Cancer vaccine development for hepatocellular carcinoma – HEPAVAC
- Author
-
Toni Weinschenk, Hans-Georg Rammensee, Danila Valmori, Roberto S. Accolla, Andrea Mayer, Regina Heidenreich, Sven Francque, Bruno Sangro, Yuk Ting Ma, Francesco Izzo, Sarah Kutscher, Harpreet Singh-Jasuja, Phillip Mueller, Markus W. Loeffler, Alfred Koenigsrainer, and Luigi Buonaguro
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Immunogenicity ,medicine.medical_treatment ,Immunology ,Cancer ,Human leukocyte antigen ,medicine.disease ,Epitope ,Cancer immunotherapy ,Internal medicine ,Hepatocellular carcinoma ,Medicine ,Cancer vaccine ,business ,Adjuvant - Abstract
The HEPAVAC Consortium aims to develop a highly innovative, novel cancer vaccine approach for hepatocellular carcinoma (HCC). The international project consortium consists of 9 European Partners from academia and the biotech industry with complementary and substantial expertise in developing immunotherapeutic strategies to treat cancer. The project has started in September 2013 and is supported by the European Commission's 7th Framework Program (www.hepavac.eu). HCC/normal adjacent tissue matched samples have been collected for HLA immunopeptidome analysis. 17 HCC samples from HLA-A*02+ patients and 15 samples from HLA-A*24+ patients have been analysed by mass spectrometry (LC-MS/MS). RNA-expression profiles have been established for 12 HCC samples. HLA-presentation/expression of peptides and mRNA on primary HCC samples are compared to n>140 normal tissue samples from relevant organs (including heart, brain, lung, kidney, liver, nerve, skin etc.) from Immatics' database. A total of 9051 HLA-A*02-restricted different tumor-associated peptides (TUMAPs) have been identified from HLA-A*02+ samples, while a total of 3286 different HLA-A*24-restricted TUMAPs have been identified from HLA-A*24+ samples. Of these, 33 HLA-A*02+ TUMAPS and 33 HLA-A*24+ TUMAPs are currently in the process of validation, including peptide synthesis, immunogenicity testing and pharmaceutical evaluation. Most promising TUMAP candidates show selective expression only in HCC samples and no expression in normal tissues. In parallel, more than 6600 HLA-DR TUMAPs have been identified in Hep3B cells transfected with CIITA as well as an average of 1500 HLA-DR TUMAPs have been identified in HCC samples. Additional HLA-DR TUMAPs derived by distinct CIITA-transfected cell lines with different HLA-DR haplotypes are under scrutiny. The discovery phase of the HEPAVAC project is proceeding according to the proposed timelines. HCC-specific epitopes to be included in the vaccine cocktail will be selected in the coming weeks for GMP production. In parallel, preclinical studies assessing the formulation and combination of the immunological RNA-based adjuvant (RNAdjuvant®) with peptide cocktails are underway. Citation Format: Luigi Buonaguro, Sarah Kutscher, Roberto Accolla, Yuk T. Ma, Regina Heidenreich, Francesco Izzo, Alfred Koenigsrainer, Markus Loeffler, Phillip Mueller, Andrea Mayer, Hans-Georg Rammensee, Bruno Sangro, Sven Francque, Danila Valmori, Toni Weinschenk, Harpreet Singh-Jasuja. Cancer vaccine development for hepatocellular carcinoma – HEPAVAC. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr A115.
- Published
- 2016
38. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival
- Author
-
Hans-Georg Rammensee, Dominik Maurer, Alexandra Kirner, Janusz J. Stanczak, Hisakazu Yamagishi, Romuald Zdrojowy, Heike Pohla, Regina Mendrzyk, Jens Fritsche, Vincenzo Bronte, Verona Vass, Susanna Mandruzzato, Natsuki Takaha, Tilo Biedermann, Michael Staehler, Jurgen Frisch, Pierre-Yves Dietrich, Steffen Walter, Arnulf Stenzl, Hiroaki Tanaka, Tsuneharu Miki, Toni Weinschenk, Andrea Mayer-Mokler, Christian Flohr, Harpreet Singh-Jasuja, Fumiya Hongo, Claudia Trautwein, Anna Pluzanska, Cezary Szczylik, Stefan Stevanovic, Peter Lewandrowski, Wolfram Brugger, Oliver Schoor, Kosei Hirakawa, Norbert Hilf, Evelyna Derhovanessian, Graham Pawelec, Andrea Mahr, Carsten Reinhardt, and Dietrich, Pierre-Yves
- Subjects
Oncology ,Male ,medicine.medical_treatment ,Kaplan-Meier Estimate ,CD8-Positive T-Lymphocytes ,T-Lymphocytes, Regulatory ,Cancer immunotherapy ,RENAL-CELL CARCINOMA ,HLA A2 antigen ,apolipoprotein B ,apolipoprotein C ,apolipoprotein A ,apolipoprotein D ,apolipoprotein E ,Antigens, Neoplasm/immunology ,ddc:616 ,Immunosuppressive Agents/administration & dosage/pharmacology/therapeutic use ,HIGH-FREQUENCIES ,FOXP3 ,General Medicine ,COLONY-STIMULATING FACTOR ,Middle Aged ,Prognosis ,Combined Modality Therapy ,Tumor antigen ,Kidney Neoplasms ,PROSTATE-CANCER ,unclassified drug ,PULSED DENDRITIC CELLS ,medicine.anatomical_structure ,Treatment Outcome ,RENAL-CELL CARCINOMA, REGULATORY T-CELLS, COLONY-STIMULATING FACTOR, MYELOID SUPPRESSOR-CELLS, PULSED DENDRITIC CELLS, TUMOR-ANTIGEN, MELANOMA PATIENTS, HIGH-FREQUENCIES, PROSTATE-CANCER, PEPTIDE VACCINE ,Vaccines, Subunit ,Chemokine CCL17/blood ,Biological Markers ,Female ,cancer vaccine ,Immunosuppressive Agents ,medicine.medical_specialty ,Cyclophosphamide/administration & dosage/pharmacology/therapeutic use ,T cell ,Apolipoprotein A-I/blood ,transcription factor FOXP3 ,ima 901 ,CD8-Positive T-Lymphocytes/immunology ,Cancer Vaccines ,General Biochemistry, Genetics and Molecular Biology ,Lymphocyte Depletion ,Cancer Vaccines/therapeutic use ,T-Lymphocytes, Regulatory/drug effects/immunology ,apolipoprotein A, apolipoprotein B, apolipoprotein C, apolipoprotein D, apolipoprotein E, cancer vaccine, cyclophosphamide, HLA A2 antigen, ima 901, thymus and activation regulated chemokine, transcription factor FOXP3, unclassified drug ,Immune system ,Antigen ,Antigens, Neoplasm ,Predictive Value of Tests ,Internal medicine ,PEPTIDE VACCINE ,HLA-A2 Antigen ,medicine ,Humans ,Vaccines, Subunit/therapeutic use ,REGULATORY T-CELLS ,TUMOR-ANTIGEN ,Carcinoma, Renal Cell/blood/drug therapy/immunology/therapy ,Carcinoma, Renal Cell ,Cyclophosphamide ,thymus and activation regulated chemokine ,Kidney Neoplasms/blood/drug therapy/immunology/therapy ,Apolipoprotein A-I ,business.industry ,Granulocyte-Macrophage Colony-Stimulating Factor ,Immunotherapy, Active ,Immunotherapy ,Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use ,MELANOMA PATIENTS ,Immunology ,MYELOID SUPPRESSOR-CELLS ,Cancer vaccine ,Chemokine CCL17 ,business ,HLA-A2 Antigen/immunology ,Biomarkers - Abstract
IMA901 is the first therapeutic vaccine for renal cell cancer (RCC) consisting of multiple tumor-associated peptides (TUMAPs) confirmed to be naturally presented in human cancer tissue. We treated a total of 96 human leukocyte antigen A (HLA-A)*02(+) subjects with advanced RCC with IMA901 in two consecutive studies. In the phase 1 study, the T cell responses of the patients to multiple TUMAPs were associated with better disease control and lower numbers of prevaccine forkhead box P3 (FOXP3)(+) regulatory T (T(reg)) cells. The randomized phase 2 trial showed that a single dose of cyclophosphamide reduced the number of T(reg) cells and confirmed that immune responses to multiple TUMAPs were associated with longer overall survival. Furthermore, among six predefined populations of myeloid-derived suppressor cells, two were prognostic for overall survival, and among over 300 serum biomarkers, we identified apolipoprotein A-I (APOA1) and chemokine (C-C motif) ligand 17 (CCL17) as being predictive for both immune response to IMA901 and overall survival. A randomized phase 3 study to determine the clinical benefit of treatment with IMA901 is ongoing.
- Published
- 2012
39. The mouse dendritic cell marker CD11c is down-regulated upon cell activation through Toll-like receptor triggering
- Author
-
Marie-Christophe Boissier, Patrice Decker, Matthieu Ribon, Natacha Bessis, Allan Thiolat, Harpreet Singh-Jasuja, and Hans-Georg Rammensee
- Subjects
Immunology ,Cell ,Down-Regulation ,chemical and pharmacologic phenomena ,Biology ,Major histocompatibility complex ,Mice ,medicine ,Immunology and Allergy ,Animals ,Humans ,CD40 Antigens ,Cells, Cultured ,CD86 ,Mice, Knockout ,Toll-like receptor ,MHC class II ,CD40 ,Toll-Like Receptors ,Histocompatibility Antigens Class II ,hemic and immune systems ,Cell Differentiation ,Hematology ,Dendritic Cells ,Molecular biology ,CD11c Antigen ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Myeloid Differentiation Factor 88 ,biology.protein ,Cytokine secretion ,Cell activation ,Biomarkers ,Signal Transduction - Abstract
Dendritic cells (DC) play a key role in regulating immune responses and are the best professional antigen-presenting cells. Two major DC populations are defined in part according to cell surface CD11c expression levels. Unexpectedly, we observed that mouse DC strongly down-regulate the typical DC marker CD11c upon activation. To better characterize DC responses, we have analyzed CD11c expression on mouse and human myeloid DC after Toll-like receptor (TLR) triggering. Here we show that mouse bone marrow-derived DC (BMDC) as well as spleen DC down-regulate cell surface CD11c upon activation by TLR3/4/9 agonists. In all cases, full DC activation was reached, as determined by cytokine secretion, cell stimulation in mixed leukocyte reactions (MLR), and CD40/CD86/major histocompatibility complex (MHC) up-regulation. Interestingly, membrane CD11c down-regulation correlated with increased cytoplasmic pools of CD11c. In contrast to the up-regulation of CD40 and MHC class II molecules, lipopolysaccharide (LPS)-induced CD11c down-regulation was MyD88-dependent. Polyinosinic-polycytidylic acid (poly I:C), which does not signal through MyD88, also induced cell surface CD11c down-regulation. Notably, CD11c down-regulation was not observed upon activation of human DC, either through TLR-dependent or -independent cell activation. Thus, activated mouse DC may be transiently CD11c-negative in vivo, hampering the identification of those cells. On the other hand, cell surface CD11c down-regulation may serve as a new activation marker for mouse DC.
- Published
- 2011
40. Defining the critical hurdles in cancer immunotherapy
- Author
-
Andrea Nicolini, Francesco M. Marincola, William E. Carson, Paolo A. Ascierto, Michele Maio, Jedd D. Wolchok, Michael T. Lotze, Jirina Bartunkova, Weihua Xiao, Hauke Winter, Barbara Seliger, Jon M. Wigginton, Cedrik M. Britten, Ignacio Melero, Guido Kroemer, Neil L. Berinstein, Jill O'Donnell-Tormey, Heinz Zwierzina, Lothar Bergmann, Lloyd J. Old, Christian H. Ottensmeier, Jérôme Galon, Per thor Straten, Koji Kawakami, Michael Papamichail, Yutaka Kawakami, Michael I. Nishimura, Mary L. Disis, Steinar Aamdal, C. J. M. Melief, Pedro Romero, Kristen Hege, Wenru Song, Pawel Kalinski, Jonathan L. Bramson, Harpreet Singh-Jasuja, Jens Peter Marschner, Bernard A. Fox, Samir N. Khleif, Brad H. Nelson, Marij J. P. Welters, Elizabeth M. Jaffee, Patrick Hwu, Rik J. Scheper, Robert C. Rees, Giuseppe Masucci, Hideaki Tahara, Cristina Bonorino, Glenn Dranoff, Ernest C. Borden, William J. Murphy, Zhigang Tian, Michael B. Atkins, Robert O. Dillman, Thomas F. Gajewski, Hiroshi Shiku, Leif Håkansson, Michael J. Mastrangelo, Lisa H. Butterfield, Shukui Qin, Laurence Zitvogel, Harry Dolstra, Michele Guida, George Coukos, Mohamed L. Salem, Xuetao Cao, Giorgio Parmiani, Enrico Proietti, Ena Wang, Sylvia Janetzki, A. Raja Choudhury, Gerd Ritter, Hyam I. Levitsky, Kunle Odunsi, Kohzoh Imai, Paul von Hoegen, Christoph Huber, Réjean Lapointe, Antoni Ribas, Dolores J. Schendel, Pamela S. Ohashi, Beatrix Kotlan, Cécile Gouttefangeas, James H. Finke, Alfred E. Chang, Howard L. Kaufman, Lindy G. Durrant, Sjoerd H. van der Burg, Jared Gollob, Dainius Characiejus, Tara Withington, Padmanee Sharma, Ronald B. Herberman, Cristina Maccalli, Ulrich Keilholtz, Axel Hoos, Graham Pawelec, Fabio Grizzi, Tanja D. de Gruijl, F. Stephen Hodi, Ruggero Ridolfi, James P. Allison, Licia Rivoltini, Carl H. June, Rolf Kiessling, Department of Molecular Microbiology and Immunology, Oregon Health and Science University [Portland] (OHSU)-Knight Cancer Institute, Earle A. Chiles Research Institute, Providence Portland Medical Center-Robert W. Franz Research Center-Providence Cancer Center, Clinical Cooperation Group 'Immune Monitoring', German Research Center for Environmental Health-Helmholtz Centre Munich-Institute of Molecular Immunology, Division of Hematology Oncology, University of Pittsburgh Cancer Institute-Departments of Medicine, Department of Surgery, Cancer Institute-University of Pittsburgh (PITT), Pennsylvania Commonwealth System of Higher Education (PCSHE)-Pennsylvania Commonwealth System of Higher Education (PCSHE), Department of Immunology, University of Pittsburgh Cancer Institute, Department of Clinical Cancer Research, The Norwegian Radium Hospital-Oslo University Hospital [Oslo], Memorial Sloane Kettering Cancer Center [New York], Howard Hughes Medical Institute (HHMI), Medical Oncology and Innovative Therapy, Instituto Nazionale Tumori-Fondazione 'G. Pascale', Beth Israel Deaconess Medical Center, Harvard Medical School [Boston] (HMS), Institute of Immunology, Charles University [Prague] (CU)-FOCIS Center of Excellence-2nd Medical School, Goethe-Universität Frankfurt am Main, IRX Therapeutics, Stanford University-ImmunoVaccine Inc., Instituto Nacional para o Controle do Câncer, Instituto de Pesquisas Biomédicas-PUCRS Faculdade de Biociências, Department of Solid Tumor Oncology, Cleveland Clinic, Department of Translational Hematology and Oncology Research, Department of Pathology, McMaster University [Hamilton, Ontario], University Medical Center Mainz, III. Medical Department, Ribological GmbH, Department of Medicine-University Medical Center of the Johannes Gutenberg-University-Clinical Development, BioNTech AG, Chinese Academy of Medical Sciences, Second Military Medical University-National Key Laboratory of Medical Immunology, Ohio State University [Columbus] (OSU), University of Michigan [Ann Arbor], University of Michigan System-University of Michigan System, Institute of Oncology, Vilnius University [Vilnius]-Faculty of Medicine, Department of Medicine, University of Queensland [Brisbane], Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania [Philadelphia]-University of Pennsylvania [Philadelphia], Department of Medical Oncology, VU Medical Center-Cancer Center Amsterdam, Hoag Institute for Research and Education, Hoag Cancer Institute, Department of Laboratory Medicine, Radboud university [Nijmegen]-Nijmegen Centre for Molecular Life Sciences-Nijmegen Medical Centre [Nijmegen], Brigham and Women's Hospital [Boston], Dana-Farber Cancer Institute [Boston], Academic Department of Clinical Oncology, University of Nottingham, UK (UON), Centre de Recherche des Cordeliers (CRC (UMR_S 872)), Université Paris Descartes - Paris 5 (UPD5)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Alnylam Pharmaceuticals, Inc., Institute for Cell Biology, Istituto Clinico Humanitas [Milan] (IRCCS Milan), Humanitas University [Milan] (Hunimed), Oncology Department, University of Lund, CanImGuide Therapeutics AB, University of California [San Francisco] (UCSF), University of California, Intrexon Corporation, Germantown, Bristol-Myers Squibb Company, Translational Oncology & Immunology, Centre TRON at the Mainz University Medical Center, Department of Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center [Houston], The Institute of Medical Science, The University of Tokyo (UTokyo), Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine [Baltimore]-Johns Hopkins University School of Medicine [Baltimore], ZellNet Consulting, Pathology and Laboratory Medicine, University of Pennsylvania [Philadelphia], Rush University Cancer Center, Rush University Medical Center [Chicago], School of Medicine and Public Health, Kyoto University [Kyoto], Division of Cellular Signaling, Institute for Advanced Medical Research, Dept. of Hematology and Medical Oncology, Charité Comprehensive Cancer Center, Cancer Vaccine Section, NCI, Department of Oncology - Pathology, Cancer Center Karolinska [Karolinska Institutet] (CCK), Karolinska Institutet [Stockholm]-Karolinska Institutet [Stockholm], Department of Molecular Immunology and Toxicology, Center of Surgical and Molecular Tumor pathology, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR CHUM), Centre Hospitalier de l'Université de Montréal (CHUM), Université de Montréal (UdeM)-Université de Montréal (UdeM), School of Medicine, Johns Hopkins University (JHU)-Oncology Center, Department of Molecular Oncology, Foundation San Raffaele Scientific Institute, Medical Oncology and Immunotherapy, Istituto Toscano Tumori-University Hospital of Siena-Department of Oncology, Merck KGaA, Merck & Co. Inc, Thomas Jefferson University, Department of Oncology-Pathology, karolinska institute, CIMA, CUN and Medical School University of Navarra, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center (LUMC), Davis Medical Center, Sacramento-University of California, Deeley Research Centre, BC Cancer Agency (BCCRC), Department of Internal Medicine, University of Pisa - Università di Pisa, Oncology Institute, Loyola University Medical Center (LUMC)-Cardinal Bernardin Cancer Center, Tumor Immunology and Immunotherapy Program, Roswell Park Cancer Institute [Buffalo]-Department of Gynecologic Oncology, Ontario Cancer Institute, University Health Network, Cancer Immunotherapy Consortium (CIC), Cancer Research Institute, Cancer Research, Ludwig Institute, Experimental Cancer Medicine Centre, University of Southampton-Faculty of Medicine, Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Unit of Immuno-Biotherapy of Melanoma and Solid Tumors, San Raffaele Scientific Institute, Center for Medical Research, Eberhard Karls Universität Tübingen = Eberhard Karls University of Tuebingen, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanita', Chinese PLA Cancer Center, Department of Oncology-The Eighty-First Hospital, The John van Geest Cancer Research Centre, School of Science and Technology-Nottingham Trent University, Jonsson Comprehensive Cancer Center, Immunoterapia e Terapia Cellulare Somatica, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.), Unit of Immunotherapy of Human Tumors, Istituto Nazionale Tumori-IRCCS Foundation, Division of Clinical Onco-Immunology, Université de Lausanne (UNIL)-Ludwig Center for Cancer Research, Immunology and Biotechnology Unit, Faculty of Science-Department of Zoology-Tanta University, VU University Medical Center [Amsterdam], Institute of Medical Immunology, Martin-Luther-Universität Halle Wittenberg (MLU), Departments of Immunology, Department of Cancer Vaccine, Mie University, Department of Immuno-gene Therapy, Immatics Biotechnologies GmbH, Eberhard Karls Universität Tübingen = Eberhard Karls University of Tuebingen-Department of Immunology-Institute for Cell Biology, Millennium: The Takeda Oncology Company, Pfizer Oncology, Center for Cancer Immune Therapy (CCIT), Herlev and Gentofte Hospital-Department of Hematology, Department of Surgery and Bioengineering, The University of Tokyo (UTokyo)-Institute of Medical Science-Advanced Clinical Research Center, School of Life Sciences-University of Science & Technology of China [Suzhou], Institute of Immunopharmacology & Immunotherapy, Shandong University-School of Pharmaceutical Sciences, Experimental Cancer Immunology and Therapy, Leiden University Medical Center (LUMC)-Department of Clinical Oncology, Euraccine Consulting Group, Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine-Clinical Center-National Institute of Health NIH), Center for Human Immunology (CHI), National Institute of Health (NIH), Leiden University Medical Center (LUMC)-Department of Clinical Oncology (K1-P), Ludwig Maximilians University-Klinikum Grosshadern, Biological Therapy of Cancer, Medical and Surgical Services Organizations-International Society For Biological Therapy Of Cancer, School of Life Science-University of Science and Technology of China [Hefei] (USTC), Immunologie des tumeurs et immunothérapie (UMR 1015), Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Gustave Roussy (IGR)-Université Paris-Sud - Paris 11 (UP11), Department Haematology and Oncology, Innsbruck Medical University [Austria] (IMU), Medical Center, University of Chicago, Discovery Medicine-Oncology, Tumor Vaccine Group, University of Washington [Seattle]-Center for Translational Medicine in Women's Health, The work of CIMT-CIP was supported by a grant from the Wallace Coulter foundation (Florida, USA)., Helmholtz Centre Munich-Institute of Molecular Immunology-Helmholtz Zentrum München = German Research Center for Environmental Health, University of Pennsylvania-University of Pennsylvania, Radboud University [Nijmegen]-Nijmegen Centre for Molecular Life Sciences-Nijmegen Medical Centre [Nijmegen], Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Lund University [Lund], University of California [San Francisco] (UC San Francisco), University of California (UC), University of Pennsylvania, Kyoto University, Sacramento-University of California (UC), Université de Lausanne = University of Lausanne (UNIL)-Ludwig Center for Cancer Research, Klinikum Grosshadern-Ludwig-Maximilians University [Munich] (LMU), University of Science and Technology of China [Hefei] (USTC)-School of Life Science, Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM), Innsbruck Medical University = Medizinische Universität Innsbruck (IMU), BMC, Ed., Computer Systems, Medical oncology laboratory, Pathology, CCA - Immuno-pathogenesis, CCA - Innovative therapy, Oregon Health and Science University-Knight Cancer Institute, Cancer Institute-University of Pittsburgh, The Norwegian Radium Hospital-Oslo University Hospital, Memorial Sloan-Kettering Cancer Center, Sloan-Kettering Institute, Howard Hughes Medical Institute, Ludwig Center for Cancer Immunotherapy, A Teaching Hospital of Harvard Medical School, Charles University [Prague]-FOCIS Center of Excellence-2nd Medical School, Stanford University [Stanford]-ImmunoVaccine Inc., Ohio State University [Columbus] ( OSU ), University of Michigan Medical Center, University of Pennsylvania Medical Center, Radboud university [Nijmegen]-Nijmegen Centre for Molecular Life Sciences-Nijmegen Medical Centre, University of Nottingham, UK ( UON ), Cleveland Clinic Foundation, Centre de Recherche des Cordeliers ( CRC (UMR_S 872) ), Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ), Istituto Clinico Humanitas [Milan] ( IRCCS Milan ), Humanitas University [Milan] ( Hunimed ), University of California [San Francisco] ( UCSF ), Harvard Medical School [Boston] ( HMS ), MD Anderson Cancer Center, The University of Tokyo, Rush University Medical Center, Cancer Center Karolinska [Karolinska Institutet] ( CCK ), Centre Hospitalier de l'Université de Montréal-Hôpital Notre-Dame Research Center ( CRCHUM ), Department of Medicine-University of Montreal, Johns Hopkins University ( JHU ) -Oncology Center, BC Cancer Agency ( BCCRC ), University of Pisa [Pisa], Loyola University Medical Center ( LUMC ) -Cardinal Bernardin Cancer Center, Cancer Immunotherapy Consortium ( CIC ), University of Southampton [Southampton]-Faculty of Medicine, Eberhard Karls Universität Tübingen, University of Lausanne-Ludwig Center for Cancer Research, Martin-Luther-University Halle-Wittenberg, Mie University Graduate School of Medicine, Eberhard Karls Universität Tübingen-Department of Immunology-Institute for Cell Biology, Center for Cancer Immune Therapy ( CCIT ), Herlev Hospital-Department of Hematology, The University of Tokyo-Institute of Medical Science-Advanced Clinical Research Center, Infectious Disease and Immunogenetics Section ( IDIS ), Center for Human Immunology ( CHI ), University of Science and Technology of China [Hefei] ( USTC ) -School of Life Science, Immunologie des tumeurs et immunothérapie ( UMR 1015 ), Université Paris-Sud - Paris 11 ( UP11 ) -Institut Gustave Roussy ( IGR ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ), Innsbruck Medical University [Austria] ( IMU ), Department of Medicine-Clinical Development, BioNTech AG-Johannes Gutenberg - Universität Mainz = Johannes Gutenberg University (JGU), Universiteit Leiden-Universiteit Leiden, Roswell Park Cancer Institute [Buffalo] (RPCI)-Department of Gynecologic Oncology, Istituto Superiore di Sanità (ISS), Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Universiteit Leiden-Universiteit Leiden-Department of Clinical Oncology, and Universiteit Leiden-Universiteit Leiden-Department of Clinical Oncology (K1-P)
- Subjects
medicine.medical_specialty ,International Cooperation ,medicine.medical_treatment ,Alternative medicine ,lcsh:Medicine ,Translational research ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Cancer Immunotherapy ,General Biochemistry, Genetics and Molecular Biology ,[ SDV.CAN ] Life Sciences [q-bio]/Cancer ,Translational Research, Biomedical ,03 medical and health sciences ,SDG 17 - Partnerships for the Goals ,0302 clinical medicine ,Cancer immunotherapy ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,[ SDV.MHEP ] Life Sciences [q-bio]/Human health and pathology ,Neoplasms ,medicine ,Humans ,In patient ,030304 developmental biology ,Medicine(all) ,0303 health sciences ,geography ,Summit ,geography.geographical_feature_category ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,Biochemistry, Genetics and Molecular Biology(all) ,business.industry ,lcsh:R ,Cancer ,General Medicine ,Public relations ,medicine.disease ,3. Good health ,Clinical trial ,Immunotherapy ,Neoplasms/therapy ,Translational Medical Research ,030220 oncology & carcinogenesis ,Immunology ,Commentary ,Working group ,business ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
Scientific discoveries that provide strong evidence of antitumor effects in preclinical models often encounter significant delays before being tested in patients with cancer. While some of these delays have a scientific basis, others do not. We need to do better. Innovative strategies need to move into early stage clinical trials as quickly as it is safe, and if successful, these therapies should efficiently obtain regulatory approval and widespread clinical application. In late 2009 and 2010 the Society for Immunotherapy of Cancer (SITC), convened an "Immunotherapy Summit" with representatives from immunotherapy organizations representing Europe, Japan, China and North America to discuss collaborations to improve development and delivery of cancer immunotherapy. One of the concepts raised by SITC and defined as critical by all parties was the need to identify hurdles that impede effective translation of cancer immunotherapy. With consensus on these hurdles, international working groups could be developed to make recommendations vetted by the participating organizations. These recommendations could then be considered by regulatory bodies, governmental and private funding agencies, pharmaceutical companies and academic institutions to facilitate changes necessary to accelerate clinical translation of novel immune-based cancer therapies. The critical hurdles identified by representatives of the collaborating organizations, now organized as the World Immunotherapy Council, are presented and discussed in this report. Some of the identified hurdles impede all investigators; others hinder investigators only in certain regions or institutions or are more relevant to specific types of immunotherapy or first-in-humans studies. Each of these hurdles can significantly delay clinical translation of promising advances in immunotherapy yet if overcome, have the potential to improve outcomes of patients with cancer. © 2011 Fox et al; licensee BioMed Central Ltd.
- Published
- 2011
41. Cancer Vaccines: Some Basic Considerations
- Author
-
Hans-Georg Rammensee, Steve Pascolo, Harpreet Singh-Jasuja, and Niels Emmerich
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,medicine ,Cancer ,medicine.disease ,business - Published
- 2009
42. Glycoprotein 96-activated dendritic cells induce a CD8-biased T cell response
- Author
-
Sabina Rayo Ramirez, Harpreet Singh-Jasuja, Katrin Wiemann, Sibylla Braedel-Ruoff, Hans-Georg Rammensee, Norbert Hilf, Hansjörg Schild, and Tobias Warger
- Subjects
CD4-Positive T-Lymphocytes ,Lipopolysaccharides ,Antigen-Presenting Cells ,Bone Marrow Cells ,Mice, Transgenic ,Receptors, Cell Surface ,Biology ,CD8-Positive T-Lymphocytes ,Major histocompatibility complex ,Lymphocyte Activation ,Biochemistry ,Mice ,Immune system ,Heat shock protein ,Cytotoxic T cell ,Animals ,Humans ,Antigen-presenting cell ,Cells, Cultured ,Membrane Glycoproteins ,Toll-Like Receptors ,Cell Differentiation ,Cell Biology ,Dendritic cell ,Dendritic Cells ,Original Articles ,Acquired immune system ,Lymphocyte Subsets ,Cell biology ,Mice, Inbred C57BL ,Toll-Like Receptor 4 ,biology.protein ,Inflammation Mediators ,CD8 ,Signal Transduction - Abstract
Heat shock proteins (Hsps) are able to induce protective immune responses against pathogens and tumors after injection into immunocompetent hosts. The activation of components of the adaptive immune system, including cytotoxic T lymphocytes specific for pathogen- or tumor-derived peptides, is crucial for the establishment of immuno- protection. Hsps acquire these peptides during intracellular protein degradation and when released during necrotic cell death, facilitate their uptake and Minor Histocompatibility Complex (MHC)-restricted representation by professional antigen-presenting cells (APCs). In addition, the interaction of Hsps with APCs, including the Endoplasmatic Reticulum (ER)-resident chaperone glycoprotein 96 (Gp96), induces the maturation of these cells by Toll-like receptor (TLR)- mediated signaling events. We now provide evidence that in contrast to lipopolysaccharides (LPS)-mediated dendritic cell (DC) maturation, the interaction of Gp96 with DCs leads to the preferential expansion of antigen-specific CD8- positive T cells in vitro and in vivo. This CD8 preference induced by mouse and human DCs did not correlate with enhanced levels of interleukin-12 secretion. Thus, despite the fact that both LPS and Gp96 activate DCs in a TLR4- dependent manner, the experiments of this study clearly demonstrate qualitative differences in the outcome of this maturation process, which preferentially favors the expansion of CD8-positive T cells.
- Published
- 2005
43. Nucleosome, the main autoantigen in systemic lupus erythematosus, induces direct dendritic cell activation via a MyD88-independent pathway: consequences on inflammation
- Author
-
Patrice Decker, Ina Kötter, Hans-Georg Rammensee, Harpreet Singh-Jasuja, and Sabine Haager
- Subjects
Immunology ,Inflammation ,Autoimmunity ,Biology ,In Vitro Techniques ,Immune complex formation ,Autoantigens ,Mice ,medicine ,Immunology and Allergy ,Animals ,Humans ,Lupus Erythematosus, Systemic ,Secretion ,Receptors, Immunologic ,Adaptor Proteins, Signal Transducing ,CD86 ,Mice, Knockout ,Mice, Inbred BALB C ,Systemic lupus erythematosus ,Peripheral tolerance ,Cell Differentiation ,Dendritic cell ,Dendritic Cells ,medicine.disease ,Antigens, Differentiation ,Nucleosomes ,Endotoxins ,Mice, Inbred C57BL ,Self Tolerance ,Myeloid Differentiation Factor 88 ,Pinocytosis ,Cytokine secretion ,Female ,medicine.symptom - Abstract
Nucleosome is the major autoantigen in systemic lupus erythematosus. It is found as a circulating complex in the sera of patients and seems to play a key role in disease development. In this study, we show for the first time that physiologic concentrations of purified nucleosomes directly induce in vitro dendritic cell (DC) maturation of mouse bone marrow-derived DC, human monocyte-derived DC (MDDC), and purified human myeloid DC as observed by stimulation of allogenic cells in MLR, cytokine secretion, and CD86 up-regulation. Importantly, nucleosomes act as free complexes without the need for immune complex formation or for the presence of unmethylated CpG DNA motifs, and we thus identified a new mechanism of DC activation by nucleosomes. We have clearly demonstrated that this activation is nucleosome-specific and endotoxin-independent. Particularly, nucleosomes induce MDDC to secrete cytokines known to be detected in high concentrations in the sera of patients. Moreover, activated MDDC secrete IL-8, a neutrophil chemoattractant also detected in patient sera, and thus might favor the inflammation observed in patients. Both normal and lupus MDDC are sensitive to nucleosome-induced activation. Finally, injection of purified nucleosomes to normal mice induces in vivo DC maturation. Altogether, these results strengthen the key role of nucleosomes in systemic lupus erythematosus and might explain how peripheral tolerance is broken in patients.
- Published
- 2005
44. The Tübingen approach: identification, selection, and validation of tumor-associated HLA peptides for cancer therapy
- Author
-
Hans-Georg Rammensee, Harpreet Singh-Jasuja, and Niels Emmerich
- Subjects
Cancer Research ,medicine.medical_treatment ,T-Lymphocytes ,Immunology ,Epitopes, T-Lymphocyte ,Context (language use) ,Human leukocyte antigen ,Computational biology ,Major histocompatibility complex ,Epitope ,Antigen ,Cancer immunotherapy ,Antigens, Neoplasm ,Neoplasms ,medicine ,Immunology and Allergy ,Humans ,biology ,Histocompatibility Antigens Class I ,Genomics ,medicine.disease ,Acquired immune system ,Primary tumor ,Oncology ,biology.protein ,Immunotherapy ,T-Lymphocytes, Cytotoxic - Abstract
There is substantial need for molecularly defined tumor antigens to prime cytotoxic T cells in vivo for cancer immunotherapy, especially in the case of tumor entities for which only a few tumor antigens have been defined so far. In this review, we present the “Tubingen approach” to identify, select, and validate large numbers of MHC/HLA class I–associated peptides derived from tumor-associated antigens. Step 1 is the identification of naturally presented HLA-associated peptides directly from primary tumor cells. Step 2 is selection of tumor-associated peptides from step 1 by differential gene expression analysis and data mining. Step 3 is validation of selected candidates by monitoring in vivo T-cell responses in the context of patient-individualized immunizations. Our approach combines methods from genomics, proteomics, bioinformatics, and T-cell immunology. The aim is to develop effective immunotherapeutics consisting of multiple tumor-associated epitopes in order to induce a broad and specific immune response against cancer cells.
- Published
- 2003
45. The heat shock protein Gp96 binds to human neutrophils and monocytes and stimulates effector functions
- Author
-
Sibylla Braedel, Hans-Georg Rammensee, Norbert Hilf, Harpreet Singh-Jasuja, Hansjoerg Schild, Markus P. Radsak, and Peter Brossart
- Subjects
Chemokine ,Lipopolysaccharide ,Neutrophils ,Phagocytosis ,Immunology ,Inflammation ,Biochemistry ,T-Lymphocytes, Regulatory ,Monocytes ,chemistry.chemical_compound ,Mice ,Antigens, Neoplasm ,Heat shock protein ,medicine ,Animals ,Humans ,Fluorescein isothiocyanate ,Innate immune system ,biology ,Monocyte ,Interleukin-8 ,Cell Biology ,Hematology ,Flow Cytometry ,Cell biology ,medicine.anatomical_structure ,chemistry ,biology.protein ,medicine.symptom ,Fluorescein-5-isothiocyanate ,Protein Binding - Abstract
The endoplasmic reticulum (ER)–resident heat shock protein Gp96 is involved in protein folding and is released into the extracellular space after necrotic cell death. In this context, Gp96 has immunostimulatory properties: it activates dendritic cells or macrophages and delivers associated peptides into the antigen presentation pathway, resulting in the induction of specific T-cell responses. The inflammatory response after necrotic tissue damage leads to the recruitment of polymorphonuclear neutrophils (PMNs) and monocytes, allowing them to make their first encounter with Gp96. We therefore investigated whether PMNs and monocytes interact with Gp96. We were able to show that PMNs and monocytes specifically bind fluorescein isothiocyanate (FITC)–conjugated Gp96. The binding of Gp96-FITC was competed by lipopolysaccharide (LPS) or fucoidan, a known inhibitor of scavenger receptors. Interestingly, the binding of LPS-FITC was also competed not only by fucoidan, but by Gp96, suggesting that LPS and Gp96 share a common receptor on PMNs. One important effector function of PMNs is the clearance of an inflammatory site by phagocytosis. We therefore assessed the influence of Gp96 on phagocytic activity using fluorochrome-labeled polystyrene beads. We found a marked enhancement of phagocytosis in the presence of Gp96 and concluded that PMNs not only bind Gp96, but are also activated by it. Additionally, Gp96-stimulated PMNs and especially monocytes release large amounts of interleukin-8, a potent neutrophil-attracting chemokine. In conclusion, we demonstrate that Gp96 specifically binds to and activates PMNs and monocytes, extending the function of Gp96 as a danger signal to additional members of the innate immune system.
- Published
- 2002
46. Human platelets express heat shock protein receptors and regulate dendritic cell maturation
- Author
-
Petra Schwarzmaier, Cécile Gouttefangeas, Hansjörg Schild, Harpreet Singh-Jasuja, Norbert Hilf, and Hans-Georg Rammensee
- Subjects
Blood Platelets ,Platelet Aggregation ,Immunology ,Antigen presentation ,Inflammation ,Receptors, Cell Surface ,Biology ,Biochemistry ,Binding, Competitive ,Proinflammatory cytokine ,Immune system ,Antigens, Neoplasm ,Heat shock protein ,medicine ,Humans ,Platelet activation ,Antigen-presenting cell ,Cells, Cultured ,Fluorescent Dyes ,Cell Differentiation ,Cell Biology ,Hematology ,Dendritic cell ,Dendritic Cells ,Platelet Activation ,Kinetics ,medicine.symptom ,Fluorescein-5-isothiocyanate - Abstract
Immunizations using the endoplasmic reticulum–resident heat shock protein Gp96 induce specific immune responses. Specificity is based on the major histocompatibility complex class I–restricted cross-presentation of Gp96-associated peptides derived from endogenous proteins. Initiation of the immune response depends on the ability of Gp96 to induce the production of proinflammatory cytokines by macrophages and dendritic cells (DCs) and of their maturation in a fashion presumably independent of associated peptide. Both events are mediated by Gp96 receptors on antigen-presenting cells. It is known that Gp96 is released from cells at necrosis induced, for example, by virus infection. Although this event supports the efficient induction of immune responses, it might also interfere with processes that are susceptible to chronic inflammation, such as wound healing after tissue damage. Therefore, Gp96-mediated stimulation of the immune system requires tight regulation. Here we show that human thrombocytes specifically interact with Gp96 and that binding of Gp96 to platelets is enhanced more than 10-fold on activation by thrombin. Gp96 interferes with neither thrombin-induced platelet activation nor platelet aggregation. However, the presence of platelets during Gp96-mediated DC activation reduces the secretion of proinflammatory cytokines and the activation of DCs. This effect is independent of soluble platelet factors and cell-to-cell contact between DCs and thrombocytes. Thus, we provide evidence for a regulatory mechanism that neutralizes Gp96 molecules systemically, especially in the blood. This effect might be of significance in wounds in which chronic inflammation and immune responses against autoantigens have to be prevented.
- Published
- 2002
47. The endoplasmic reticulum-resident heat shock protein Gp96 activates dendritic cells via the Toll-like receptor 2/4 pathway
- Author
-
Ramunas M. Vabulas, Sylvia Herter, Parvis Ahmad-Nejad, Hans-Georg Rammensee, Sibylla Braedel, Hermann Wagner, Carsten J. Kirschning, Norbert Hilf, Clarissa Prazeres da Costa, Harpreet Singh-Jasuja, and Hansjoerg Schild
- Subjects
Antigen presentation ,Receptors, Cell Surface ,Biology ,Major histocompatibility complex ,Endoplasmic Reticulum ,Biochemistry ,Cell Line ,Mice ,Immune system ,Animals ,Drosophila Proteins ,Humans ,Molecular Biology ,Heat-Shock Proteins ,DNA Primers ,Mice, Knockout ,Toll-like receptor ,Mice, Inbred C3H ,Innate immune system ,Membrane Glycoproteins ,Base Sequence ,Toll-Like Receptors ,Cell Biology ,Dendritic Cells ,Acquired immune system ,Toll-Like Receptor 2 ,Cell biology ,Toll-Like Receptor 4 ,TLR2 ,biology.protein ,Signal transduction ,Signal Transduction - Abstract
The heat shock protein Gp96 has been shown to induce specific immune responses. On one hand, this phenomenon is based on the specific interaction with CD91 that mediates endocytosis and results in major histocompatibility complex class I-restricted representation of the Gp96-associated peptides. On the other hand, Gp96 induces activation of professional antigen-presenting cells, resulting in the production of pro-inflammatory cytokines and up-regulation of costimulatory molecules by unknown mechanisms. In this study, we have analyzed the consequences of Gp96 interaction with cells expressing different Toll-like receptors (TLRs) and with bone marrow-derived dendritic cells from mice lacking functional TLR2 and/or TLR4 molecules. We find that the Gp96-TLR2/4 interaction results in activation of nuclear factor kappaB-driven reporter genes and mitogen- and stress-activated protein kinases and induces IkappaBalpha degradation. Bone marrow-derived dendritic cells of C3H/HeJ and more pronounced C3H/HeJ/TLR2(-/-) mice fail to respond to Gp96. Interestingly, activation of bone marrow-derived dendritic cells depends on endocytosis of Gp96 molecules. Our results provide, for the first time, the molecular basis for understanding the Gp96-mediated activation of antigen-presenting cells by describing the simultaneous stimulation of the innate and adaptive immune system. This feature explains the remarkable ability of Gp96 to induce specific immune responses against tumors and pathogens.
- Published
- 2002
48. The role of heat shock proteins and their receptors in the activation of the immune system
- Author
-
Danièle Arnold-Schild, Hansjörg Schild, Norbert Hilf, and Harpreet Singh-Jasuja
- Subjects
endocrine system ,Clinical Biochemistry ,Antigen-Presenting Cells ,chemical and pharmacologic phenomena ,Apoptosis ,Biochemistry ,Immune system ,Heat shock protein ,Cytotoxic T cell ,Animals ,Humans ,Heat shock ,Antigen-presenting cell ,Receptor ,Molecular Biology ,Heat-Shock Proteins ,Antigen processing ,Chemistry ,hemic and immune systems ,Dendritic Cells ,Cell biology ,Heat shock factor ,Immune System ,biological sciences ,Peptides ,Protein Binding - Abstract
Heat shock proteins (HSPs) have been described as potent tumor vaccines in animal models and are currently studied in clinical trials. The underlying immune response relies on immunogenic peptides that the HSPs have acquired intracellularly by interfering with the classical antigen processing pathways. There have been numerous reports shedding light on how HSPs are able to gain this function and a number of important requirements for HSP-mediated specific immunity have been described: first, the ability of HSPs to bind immunogenic peptides. Second, the acquisition of HSPs by specialized antigen presenting cells with efficient antigen processing pathways capable of inducing cellular immune responses. Third, the existence of specific receptors on the surfaces of antigen presenting cells, allowing efficient and rapid uptake of HSP-peptide complexes from the extracellular fluid. And fourth, the ability of heat shock proteins to activate antigen presenting cells, enabling the latter to prime cytotoxic T cell responses against the peptides associated to HSPs.
- Published
- 2001
49. The heat shock protein gp96: a receptor-targeted cross-priming carrier and activator of dendritic cells
- Author
-
Hansjörg Schild, Harpreet Singh-Jasuja, Hans Scherer, Hans-Georg Rammensee, René E. M. Toes, Danièle Arnold-Schild, and Norbert Hilf
- Subjects
Immunoglobulins ,chemical and pharmacologic phenomena ,Biology ,Biochemistry ,Antigens, CD ,Antigens, Neoplasm ,Heat shock protein ,Cytotoxic T cell ,Animals ,Humans ,Receptor ,Antigen-presenting cell ,CD86 ,MHC class II ,Membrane Glycoproteins ,Follicular dendritic cells ,hemic and immune systems ,Cell Biology ,Original Articles ,Dendritic Cells ,Molecular biology ,Cell biology ,CTL ,biology.protein ,B7-2 Antigen ,T-Lymphocytes, Cytotoxic - Abstract
Heat shock proteins like gp96 (grp94) are able to induce specific cytotoxic T-cell (CTL) responses against cells from which they originate and are currently studied in clinical trials for use in immunotherapy of tumors. We have recently demonstrated that gp96 binds to at least one yet unidentified receptor restricted to antigen-presenting cells (APCs) like dendritic cells (DCs) but not to T cells. Moreover we have shown, that for CTL activation by gp96-chaperoned peptides receptor-mediated uptake of gp96 by APCs is required. Lately, we have discovered a second function of gp96 when interacting with professional APCs. Gp96 is able to mediate maturation of DCs as determined by upregulation of MHC class II, CD86 and CD83 molecules, secretion of pro-inflammatory cytokines IL-12 and TNF-alpha and enhanced T-cell simulatory capacity. Furthermore, the gp96 receptor(s) are down-regulated on mature DCs, suggesting that the gp96 receptor(s) behave similar to other endocytic receptors like CD36, mannose receptor etc. Our findings now provide additional evidence for the remarkable immunogenicity of gp96: first, the existence of specific gp96 receptors on APCs and second, the capacity to activate dendritic cells which is strictly required to enable these highly sophisticated APCs to prime CTL responses.
- Published
- 2000
50. Abstract 3971: Impact of various first- and second-generation tyrosine-kinase inhibitors on frequency and functionality of immune cells
- Author
-
Harpreet Singh-Jasuja, Helen Hoerzer, Norbert Hilf, and Nina Pawlowski
- Subjects
Cancer Research ,Tivozanib ,business.industry ,Sunitinib ,medicine.medical_treatment ,FOXP3 ,Immunotherapy ,Pharmacology ,Pazopanib ,Axitinib ,Oncology ,Medicine ,Cytokine secretion ,IL-2 receptor ,business ,medicine.drug - Abstract
Combining immunotherapeutic approaches with targeted therapies like tyrosine-kinase inhibitors (TKIs) may give additional benefits to patients with VEGF-driven tumors (e.g. RCC). Previously, we have investigated the impact of first-generation TKIs sorafenib and sunitinib demonstrating distinct, in part opposing effects on immune cells (Hipp et al, Blood 2008, 111(12): 5610ff). Meanwhile, second-generation TKIs (pazopanib, axitinib, tivozanib) were approved or are expected to receive approval soon. In contrast to the multi-targeting first-generation TKIs, these TKIs act more selectively, therefore potentially inducing less off-target toxicities. However, so far little is known about the immunomodulatory properties of these new TKIs. Thus, we compared the influence of two first- and three second-generation TKIs on frequency and function of immune cells and on immune responses in mice. C57BL/6 mice were orally treated with vehicle, 25 mg/kg axitinib, 20 mg/kg tivozanib, 30 mg/kg pazopanib, 30 mg/kg sorafenib or 40 mg/kg sunitinib (daily for one week). Twelve hours after the last application, splenocytes were analyzed for CD4+ CD25+ FoxP3+ regulatory T cells (Tregs). Tivozanib and (as published before) sunitinib significantly reduced the frequency of Tregs among CD4+ cells, while no effect was observed for axitinib, pazopanib and sorafenib. Moreover, percentages of CD4+ as well as CD8+ T cells were increased in tivozanib- and sunitinib-treated mice. The functional status of isolated immune cells from TKI pre-treated mice was assessed in a mixed lymphocyte reaction (MLR). CD4+ T-cell proliferation of cells isolated from sorafenib-treated mice was diminished, while CD8+ T-cell proliferation of cells from sunitinib-treated mice was increased. In vitro activation assays revealed that - irrespective of the used TKI - the treatment led to reduced responsiveness of immune cells in terms of cytokine secretion and expression of surface activation markers. Finally, C57BL/6 mice were immunized subcutaneously twice (days 1 and 8) with ovalbumin peptide vaccine alone (negative control) or combined with poly-IC (positive control). To analyze the effect of TKIs in vivo, mice were pretreated (day -7 to -3) or concurrently treated (day 1 - 13) with axitinib, tivozanib or pazopanib in addition to vaccinations with peptide plus poly-IC. On day 15, induction of peptide-specific T cells was analyzed by MHC multimer staining as well as by IFN-γ ELISPOT. Pretreatment or concurrent treatment with all three second-generation TKIs did not negatively impact the induction of immune responses - in contrast to concurrent treatment with sorafenib reported before. In conclusion, these results suggest that immunotherapy and second-generation TKIs can be combined in the treatment of cancer, but the influence of these TKIs on the immune system requires further assessment in clinical trials. Citation Format: Nina Pawlowski, Helen Hoerzer, Harpreet Singh-Jasuja, Norbert Hilf. Impact of various first- and second-generation tyrosine-kinase inhibitors on frequency and functionality of immune cells. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 3971. doi:10.1158/1538-7445.AM2013-3971
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.