Stanford Advisors


All Publications


  • Prevalence of COVID-19-associated symptoms during acute infection in relation to SARS-CoV-2-directed humoral and cellular immune responses in a mild-diseased convalescent cohort. International journal of infectious diseases : IJID : official publication of the International Society for Infectious Diseases Tegeler, C. M., Bilich, T., Maringer, Y., Salih, H. R., Walz, J. S., Nelde, A., Heitmann, J. S. 2022

    Abstract

    OBJECTIVE: Besides SARS-CoV-2-directed humoral immune responses, T cell responses are indispensable for effective anti-viral immunity. Recent data have shown a correlation of COVID-19 symptoms with humoral immune response, but so far little is known on the association of SARS-CoV-2-directed T cell responses and disease severity. We here evaluated the prevalence of different clinical COVID-19 symptoms in relation to SARS-CoV-2-directed humoral and cellular immune responses.METHODS: Severity of eight different symptoms during acute infection were assessed questionnaire-based from 193 convalescent individuals and evaluated in relation to SARS-CoV-2 antibody levels and intensity of SARS-CoV-2-specific T cell responses 2 - 8 weeks after positive PCR.RESULTS: Whereas increased IgG serum levels could be associated with severity of most symptoms, no difference in T cell response intensity between different symptom severities was observed for the majority of COVID-19 symptoms. However, when analyzing loss of smell or taste and cough, awareness of more severe symptoms was associated with reduced T cell response intensities.CONCLUSION: These data suggest that rapid virus clearance mediated by SARS-CoV-2-specific T cells prevents severe symptoms of COVID-19.

    View details for DOI 10.1016/j.ijid.2022.04.019

    View details for PubMedID 35429640

  • Platelet PD-L1 reflects collective intratumoral PD-L1 expression and predicts immunotherapy response in non-small cell lung cancer NATURE COMMUNICATIONS Hinterleitner, C., Straehle, J., Malenke, E., Hinterleitner, M., Henning, M., Seehawer, M., Bilich, T., Heitmann, J., Lutz, M., Mattern, S., Scheuermann, S., Horger, M., Maurer, S., Walz, J., Fend, F., Handgretinger, R., Seitz, C., Weigelin, B., Singer, S., Salih, H., Borst, O., Kopp, H., Zender, L. 2021; 12 (1): 7005

    Abstract

    Immune-checkpoint inhibitors (ICI) have transformed oncological therapy. Up to 20% of all non-small cell lung cancers (NSCLCs) show durable responses upon treatment with ICI, however, robust markers to predict therapy response are missing. Here we show that blood platelets interact with lung cancer cells and that PD-L1 protein is transferred from tumor cells to platelets in a fibronectin 1, integrin α5β1 and GPIbα-dependent manner. Platelets from NSCLC patients are found to express PD-L1 and platelet PD-L1 possess the ability to inhibit CD4 and CD8 T-cells. An algorithm is developed to calculate the activation independent adjusted PD-L1 payload of platelets (pPD-L1Adj.), which is found to be superior in predicting the response towards ICI as compared to standard histological PD-L1 quantification on tumor biopsies. Our data suggest that platelet PD-L1 reflects the collective tumor PD-L1 expression, plays important roles in tumor immune evasion and overcomes limitations of histological quantification of often heterogeneous intratumoral PD-L1 expression.

    View details for DOI 10.1038/s41467-021-27303-7

    View details for Web of Science ID 000725093800011

    View details for PubMedID 34853305

    View details for PubMedCentralID PMC8636618

  • A COVID-19 peptide vaccine for the induction of SARS-CoV-2 T cell immunity NATURE Heitmann, J. S., Bilich, T., Tandler, C., Nelde, A., Maringer, Y., Marconato, M., Reusch, J., Jaeger, S., Denk, M., Richter, M., Anton, L., Weber, L., Roerden, M., Bauer, J., Rieth, J., Wacker, M., Hoerber, S., Peter, A., Meisner, C., Fischer, I., Loeffler, M. W., Karbach, J., Jaeger, E., Klein, R., Rammensee, H., Salih, H. R., Walz, J. S. 2021: 617-622

    Abstract

    T cell immunity is central for the control of viral infections. CoVac-1 is a peptide-based vaccine candidate, composed of SARS-CoV-2 T cell epitopes derived from various viral proteins1,2, combined with the Toll-like receptor 1/2 agonist XS15 emulsified in Montanide ISA51 VG, aiming to induce profound SARS-CoV-2 T cell immunity to combat COVID-19. Here we conducted a phase I open-label trial, recruiting 36 participants aged 18-80 years, who received a single subcutaneous CoVac-1 vaccination. The primary end point was safety analysed until day 56. Immunogenicity in terms of CoVac-1-induced T cell response was analysed as the main secondary end point until day 28 and in the follow-up until month 3. No serious adverse events and no grade 4 adverse events were observed. Expected local granuloma formation was observed in all study participants, whereas systemic reactogenicity was absent or mild. SARS-CoV-2-specific T cell responses targeting multiple vaccine peptides were induced in all study participants, mediated by multifunctional T helper 1 CD4+ and CD8+ T cells. CoVac-1-induced IFNγ T cell responses persisted in the follow-up analyses and surpassed those detected after SARS-CoV-2 infection as well as after vaccination with approved vaccines. Furthermore, vaccine-induced T cell responses were unaffected by current SARS-CoV-2 variants of concern. Together, CoVac-1 showed a favourable safety profile and induced broad, potent and variant of concern-independent T cell responses, supporting the presently ongoing evaluation in a phase II trial for patients with B cell or antibody deficiency.

    View details for DOI 10.1038/s41586-021-04232-5

    View details for Web of Science ID 000744417800001

    View details for PubMedID 34814158

  • SARS-CoV-2-reactive T-cell receptors isolated from convalescent COVID-19 patients confer potent T-cell effector function EUROPEAN JOURNAL OF IMMUNOLOGY Brunk, F., Moritz, A., Nelde, A., Bilich, T., Casadei, N., Fraschka, S. K., Heitmann, J. S., Hoerber, S., Peter, A., Rammensee, H., Singh, H., Walz, J., Maurer, D., Wagner, C. 2021

    Abstract

    Both B cells and T cells are involved in an effective immune response to SARS-CoV-2, the disease-causing virus of COVID-19. While B cells-with the indispensable help of CD4+ T cells-are essential to generate neutralizing antibodies, T cells on their own have been recognized as another major player in effective anti-SARS-CoV-2 immunity. In this report, we provide insights into the characteristics of individual HLA-A*02:01- and HLA-A*24:02-restricted SARS-CoV-2-reactive TCRs, isolated from convalescent COVID-19 patients. We observed that SARS-CoV-2-reactive T-cell populations were clearly detectable in convalescent samples and that TCRs isolated from these T cell clones were highly functional upon ectopic re-expression. The SARS-CoV-2-reactive TCRs described in this report mediated potent TCR signaling in reporter assays with low nanomolar EC50 values. We further demonstrate that these SARS-CoV-2-reactive TCRs conferred powerful T-cell effector function to primary CD8+ T cells as evident by a robust anti-SARS-CoV-2 IFN-γ response and in vitro cytotoxicity. We also provide an example of a long-lasting anti-SARS-CoV-2 memory response by reisolation of one of the retrieved TCRs 5 months after initial sampling. Taken together, these findings contribute to a better understanding of anti-SARS-CoV-2 T-cell immunity and may contribute to paving the way toward immunotherapeutics approaches targeting SARS-CoV-2.

    View details for DOI 10.1002/eji.202149290

    View details for Web of Science ID 000695334300001

    View details for PubMedID 34424997

  • Preexisting and Post-COVID-19 Immune Responses to SARS-CoV-2 in Patients with Cancer CANCER DISCOVERY Bilich, T., Roerden, M., Maringer, Y., Nelde, A., Heitmann, J. S., Dubbelaar, M. L., Peter, A., Hoerber, S., Bauer, J., Rieth, J., Wacker, M., Berner, F., Flatz, L., Held, S., Brossart, P., Maerklin, M., Wagner, P., Erne, E., Klein, R., Rammensee, H., Salih, H. R., Walz, J. S. 2021; 11 (8): 1982-1995

    Abstract

    Patients with cancer, in particular patients with hematologic malignancies, are at increased risk for critical illness upon COVID-19. We here assessed antibody as well as CD4+ and CD8+ T-cell responses in unexposed and SARS-CoV-2-infected patients with cancer to characterize SARS-CoV-2 immunity and to identify immunologic parameters contributing to COVID-19 outcome. Unexposed patients with hematologic malignancies presented with reduced prevalence of preexisting SARS-CoV-2 cross-reactive CD4+ T-cell responses and signs of T-cell exhaustion compared with patients with solid tumors and healthy volunteers. Whereas SARS-CoV-2 antibody responses did not differ between patients with COVID-19 and cancer and healthy volunteers, intensity, expandability, and diversity of SARS-CoV-2 T-cell responses were profoundly reduced in patients with cancer, and the latter associated with a severe course of COVID-19. This identifies impaired SARS-CoV-2 T-cell immunity as a potential determinant for dismal outcome of COVID-19 in patients with cancer. SIGNIFICANCE: This first comprehensive analysis of SARS-CoV-2 immune responses in patients with cancer reports on the potential implications of impaired SARS-CoV-2 T-cell responses for understanding pathophysiology and predicting severity of COVID-19, which in turn might allow for the development of therapeutic measures and vaccines for this vulnerable patient population.See related commentary by Salomé and Horowitz, p. 1877.This article is highlighted in the In This Issue feature, p. 1861.

    View details for Web of Science ID 000683016500029

    View details for PubMedID 34011563

  • Immunopeptidomics-Guided Warehouse Design for Peptide-Based Immunotherapy in Chronic Lymphocytic Leukemia FRONTIERS IN IMMUNOLOGY Nelde, A., Maringer, Y., Bilich, T., Salih, H. R., Roerden, M., Heitmann, J. S., Marcu, A., Bauer, J., Neidert, M. C., Denzlinger, C., Illerhaus, G., Aulitzky, W., Rammensee, H., Walz, J. S. 2021; 12: 705974

    Abstract

    Antigen-specific immunotherapies, in particular peptide vaccines, depend on the recognition of naturally presented antigens derived from mutated and unmutated gene products on human leukocyte antigens, and represent a promising low-side-effect concept for cancer treatment. So far, the broad application of peptide vaccines in cancer patients is hampered by challenges of time- and cost-intensive personalized vaccine design, and the lack of neoepitopes from tumor-specific mutations, especially in low-mutational burden malignancies. In this study, we developed an immunopeptidome-guided workflow for the design of tumor-associated off-the-shelf peptide warehouses for broadly applicable personalized therapeutics. Comparative mass spectrometry-based immunopeptidome analyses of primary chronic lymphocytic leukemia (CLL) samples, as representative example of low-mutational burden tumor entities, and a dataset of benign tissue samples enabled the identification of high-frequent non-mutated CLL-associated antigens. These antigens were further shown to be recognized by pre-existing and de novo induced T cells in CLL patients and healthy volunteers, and were evaluated as pre-manufactured warehouse for the construction of personalized multi-peptide vaccines in a first clinical trial for CLL (NCT04688385). This workflow for the design of peptide warehouses is easily transferable to other tumor entities and can provide the foundation for the development of broad personalized T cell-based immunotherapy approaches.

    View details for DOI 10.3389/fimmu.2021.705974

    View details for Web of Science ID 000675582100001

    View details for PubMedID 34305947

    View details for PubMedCentralID PMC8297687

  • T cell and antibody kinetics delineate SARS-CoV-2 peptides mediating long-term immune responses in COVID-19 convalescent individuals SCIENCE TRANSLATIONAL MEDICINE Bilich, T., Nelde, A., Heitmann, J. S., Maringer, Y., Roerden, M., Bauer, J., Rieth, J., Wacker, M., Peter, A., Hoerber, S., Rachfalski, D., Maerklin, M., Stevanovic, S., Rammensee, H., Salih, H. R., Walz, J. S. 2021; 13 (590)

    Abstract

    Long-term immunological memory to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is crucial for the development of population-level immunity, which is the aim of vaccination approaches. Reports on rapidly decreasing antibody titers have led to questions regarding the efficacy of humoral immunity alone. The relevance of T cell memory after coronavirus disease 2019 (COVID-19) remains unclear. Here, we investigated SARS-CoV-2 antibody and T cell responses in matched samples of COVID-19 convalescent individuals up to 6 months after infection. Longitudinal analysis revealed decreasing and stable spike- and nucleocapsid-specific antibody responses, respectively. In contrast, functional T cell responses remained robust, and even increased, in both frequency and intensity. Single peptide mapping of T cell diversity over time identified open reading frame-independent, dominant T cell epitopes mediating long-term SARS-CoV-2 T cell responses. Identification of these epitopes may be fundamental for COVID-19 vaccine design.

    View details for DOI 10.1126/scitranslmed.abf7517

    View details for Web of Science ID 000642350100005

    View details for PubMedID 33723016

    View details for PubMedCentralID PMC8128286

  • SARS-CoV-2-derived peptides define heterologous and COVID-19-induced T cell recognition NATURE IMMUNOLOGY Nelde, A., Bilich, T., Heitmann, J. S., Maringer, Y., Salih, H. R., Roerden, M., Luebke, M., Bauer, J., Rieth, J., Wacker, M., Peter, A., Hoerber, S., Traenkle, B., Kaiser, P. D., Rothbauer, U., Becker, M., Junker, D., Krause, G., Strengert, M., Schneiderhan-Marra, N., Templin, M. F., Joos, T. O., Kowalewski, D. J., Stos-Zweifel, V., Fehr, M., Rabsteyn, A., Mirakaj, V., Karbach, J., Jaeger, E., Graf, M., Gruber, L., Rachfalski, D., Preuss, B., Hagelstein, I., Maerklin, M., Bakchoul, T., Gouttefangeas, C., Kohlbacher, O., Klein, R., Stevanovic, S., Rammensee, H., Walz, J. S. 2021; 22 (1): 74-85

    Abstract

    T cell immunity is central for the control of viral infections. To characterize T cell immunity, but also for the development of vaccines, identification of exact viral T cell epitopes is fundamental. Here we identify and characterize multiple dominant and subdominant SARS-CoV-2 HLA class I and HLA-DR peptides as potential T cell epitopes in COVID-19 convalescent and unexposed individuals. SARS-CoV-2-specific peptides enabled detection of post-infectious T cell immunity, even in seronegative convalescent individuals. Cross-reactive SARS-CoV-2 peptides revealed pre-existing T cell responses in 81% of unexposed individuals and validated similarity with common cold coronaviruses, providing a functional basis for heterologous immunity in SARS-CoV-2 infection. Diversity of SARS-CoV-2 T cell responses was associated with mild symptoms of COVID-19, providing evidence that immunity requires recognition of multiple epitopes. Together, the proposed SARS-CoV-2 T cell epitopes enable identification of heterologous and post-infectious T cell immunity and facilitate development of diagnostic, preventive and therapeutic measures for COVID-19.

    View details for DOI 10.1038/s41590-020-00808-x

    View details for Web of Science ID 000574099400001

    View details for PubMedID 32999467

  • Mass spectrometry-based identification of a B-cell maturation antigen-derived T-cell epitope for antigen-specific immunotherapy of multiple myeloma BLOOD CANCER JOURNAL Bilich, T., Nelde, A., Bauer, J., Walz, S., Roerden, M., Salih, H. R., Weisel, K., Besemer, B., Marcu, A., Luebke, M., Schuhmacher, J., Neidert, M. C., Rammensee, H., Stevanovic, S., Walz, J. S. 2020; 10 (2): 24

    Abstract

    The B-cell maturation antigen (BCMA) is currently being evaluated as promising tumor-associated surface antigen for T-cell-based immunotherapy approaches, such as CAR T cells and bispecific antibodies, in multiple myeloma (MM). Cytotoxic T cells bearing BCMA-specific T-cell receptors might further allow targeting HLA-presented antigens derived from the intracellular domain of BCMA. By analyzing a mass spectrometry-acquired immunopeptidome dataset of primary MM samples and MM cell lines for BCMA-derived HLA ligands, we identified the naturally presented HLA-B*18-restricted ligand P(BCMA)B*18. Additionally, P(BCMA)B*18 was identified on primary CLL samples, thereby expanding the range for possible applications. P(BCMA)B*18 induced multifunctional BCMA-specific cells de novo from naïve CD8+ T cells of healthy volunteers. These T cells exhibited antigen-specific lysis of autologous peptide-loaded cells. Even in the immunosuppressive context of MM, we detected spontaneous memory T-cell responses against P(BCMA)B*18 in patients. By applying CTLA-4 and PD-1 inhibition in vitro we induced multifunctional P(BCMA)B*18-specific CD8+ T cells in MM patients lacking preexisting BCMA-directed immune responses. Finally, we could show antigen-specific lysis of autologous peptide-loaded target cells and even MM.1S cells naturally presenting P(BCMA)B*18 using patient-derived P(BCMA)B*18-specific T cells. Hence, this BCMA-derived T-cell epitope represents a promising target for T-cell-based immunotherapy and monitoring following immunotherapy in B-cell malignancy patients.

    View details for DOI 10.1038/s41408-020-0288-3

    View details for Web of Science ID 000517060200001

    View details for PubMedID 32111817

    View details for PubMedCentralID PMC7048774

  • Antigen Targets for the Development of Immunotherapies in Leukemia INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES Bauer, J., Nelde, A., Bilich, T., Walz, J. S. 2019; 20 (6)

    Abstract

    Immunotherapeutic approaches, including allogeneic stem cell transplantation and donor lymphocyte infusion, have significantly improved the prognosis of leukemia patients. Further efforts are now focusing on the development of immunotherapies that are able to target leukemic cells more specifically, comprising monoclonal antibodies, chimeric antigen receptor (CAR) T cells, and dendritic cell- or peptide-based vaccination strategies. One main prerequisite for such antigen-specific approaches is the selection of suitable target structures on leukemic cells. In general, the targets for anti-cancer immunotherapies can be divided into two groups: (1) T-cell epitopes relying on the presentation of peptides via human leukocyte antigen (HLA) molecules and (2) surface structures, which are HLA-independently expressed on cancer cells. This review discusses the most promising tumor antigens as well as the underlying discovery and selection strategies for the development of anti-leukemia immunotherapies.

    View details for DOI 10.3390/ijms20061397

    View details for Web of Science ID 000465523400037

    View details for PubMedID 30897713

    View details for PubMedCentralID PMC6471800

  • The HLA ligandome landscape of chronic myeloid leukemia delineates novel T-cell epitopes for immunotherapy BLOOD Bilich, T., Nelde, A., Bichmann, L., Roerden, M., Salih, H. R., Kowalewski, D. J., Schuster, H., Tsou, C., Marcu, A., Neidert, M. C., Luebke, M., Rieth, J., Schemionek, M., Bruemmendorf, T. H., Vucinic, V., Niederwieser, D., Bauer, J., Maerklin, M., Peper, J. K., Klein, R., Kohlbacher, O., Kanz, L., Rammensee, H., Stevanovic, S., Walz, J. S. 2019; 133 (6): 550-565

    Abstract

    Antileukemia immunity plays an important role in disease control and maintenance of tyrosine kinase inhibitor (TKI)-free remission in chronic myeloid leukemia (CML). Thus, antigen-specific immunotherapy holds promise for strengthening immune control in CML but requires the identification of CML-associated targets. In this study, we used a mass spectrometry-based approach to identify naturally presented HLA class I- and class II-restricted peptides in primary CML samples. Comparative HLA ligandome profiling using a comprehensive dataset of different hematological benign specimens and samples from CML patients in deep molecular remission delineated a panel of novel frequently presented CML-exclusive peptides. These nonmutated target antigens are of particular relevance because our extensive data-mining approach suggests the absence of naturally presented BCR-ABL- and ABL-BCR-derived HLA-restricted peptides and the lack of frequent tumor-exclusive presentation of known cancer/testis and leukemia-associated antigens. Functional characterization revealed spontaneous T-cell responses against the newly identified CML-associated peptides in CML patient samples and their ability to induce multifunctional and cytotoxic antigen-specific T cells de novo in samples from healthy volunteers and CML patients. Thus, these antigens are prime candidates for T-cell-based immunotherapeutic approaches that may prolong TKI-free survival and even mediate cure of CML patients.

    View details for DOI 10.1182/blood-2018-07-866830

    View details for Web of Science ID 000458018000009

    View details for PubMedID 30530751

  • The immunopeptidomic landscape of ovarian carcinomas PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Schuster, H., Peper, J. K., Boesmueller, H., Roehle, K., Backert, L., Bilich, T., Ney, B., Loeffler, M. W., Kowalewski, D. J., Trautwein, N., Rabsteyn, A., Engler, T., Braun, S., Haen, S. P., Walz, J. S., Schmid-Horch, B., Brucker, S. Y., Wallwiener, D., Kohlbacher, O., Fend, F., Rammensee, H., Stevanovic, S., Staebler, A., Wagner, P. 2017; 114 (46): E9942-E9951

    Abstract

    Immunotherapies, particularly checkpoint inhibitors, have set off a revolution in cancer therapy by releasing the power of the immune system. However, only little is known about the antigens that are essentially presented on cancer cells, capable of exposing them to immune cells. Large-scale HLA ligandome analysis has enabled us to exhaustively characterize the immunopeptidomic landscape of epithelial ovarian cancers (EOCs). Additional comparative profiling with the immunopeptidome of a variety of benign sources has unveiled a multitude of ovarian cancer antigens (MUC16, MSLN, LGALS1, IDO1, KLK10) to be presented by HLA class I and class II molecules exclusively on ovarian cancer cells. Most strikingly, ligands derived from mucin 16 and mesothelin, a molecular axis of prognostic importance in EOC, are prominent in a majority of patients. Differential gene-expression analysis has allowed us to confirm the relevance of these targets for EOC and further provided important insights into the relationship between gene transcript levels and HLA ligand presentation.

    View details for DOI 10.1073/pnas.1707658114

    View details for Web of Science ID 000415173300028

    View details for PubMedID 29093164

    View details for PubMedCentralID PMC5699044

  • CXCR1 Regulates Pulmonary Anti-Pseudomonas Host Defense JOURNAL OF INNATE IMMUNITY Carevic, M., Oez, H., Fuchs, K., Laval, J., Schroth, C., Frey, N., Hector, A., Bilich, T., Haug, M., Schmidt, A., Autenrieth, S. E., Bucher, K., Beer-Hammer, S., Gaggar, A., Kneilling, M., Benarafa, C., Gao, J. L., Murphy, P. M., Schwarz, S., Moepps, B., Hartl, D. 2016; 8 (4): 362-373

    Abstract

    Pseudomonas aeruginosa is a key opportunistic pathogen causing disease in cystic fibrosis (CF) and other lung diseases such as chronic obstructive pulmonary disease (COPD). However, the pulmonary host defense mechanisms regulating anti-P. aeruginosa immunity remain incompletely understood. Here we demonstrate, by studying an airway P. aeruginosa infection model, in vivo bioluminescence imaging, neutrophil effector responses and human airway samples, that the chemokine receptor CXCR1 regulates pulmonary host defense against P. aeruginosa. Mechanistically, CXCR1 regulates anti-Pseudomonas neutrophil responses through modulation of reactive oxygen species and interference with Toll-like receptor 5 expression. These studies define CXCR1 as a novel, noncanonical chemokine receptor that regulates pulmonary anti-Pseudomonas host defense with broad implications for CF, COPD and other infectious lung diseases.

    View details for DOI 10.1159/000444125

    View details for Web of Science ID 000378797300004

    View details for PubMedID 26950764

    View details for PubMedCentralID PMC4885792

  • The antigenic landscape of multiple myeloma: mass spectrometry (re) defines targets for T-cell-based immunotherapy BLOOD Walz, S., Stickel, J. S., Kowalewski, D., Schuster, H., Weisel, K., Backert, L., Kahn, S., Nelde, A., Stroh, T., Handel, M., Kohlbacher, O., Kanz, L., Salih, H., Rammensee, H., Stevanovic, S. 2015; 126 (10): 1203-1213

    Abstract

    Direct analysis of HLA-presented antigens by mass spectrometry provides a comprehensive view on the antigenic landscape of different tissues/malignancies and enables the identification of novel, pathophysiologically relevant T-cell epitopes. Here, we present a systematic and comparative study of the HLA class I and II presented, nonmutant antigenome of multiple myeloma (MM). Quantification of HLA surface expression revealed elevated HLA molecule counts on malignant plasma cells compared with normal B cells, excluding relevant HLA downregulation in MM. Analyzing the presentation of established myeloma-associated T-cell antigens on the HLA ligandome level, we found a substantial proportion of antigens to be only infrequently presented on primary myelomas or to display suboptimal degrees of myeloma specificity. However, unsupervised analysis of our extensive HLA ligand data set delineated a panel of 58 highly specific myeloma-associated antigens (including multiple myeloma SET domain containing protein) which are characterized by frequent and exclusive presentation on myeloma samples. Functional characterization of these target antigens revealed peptide-specific, preexisting CD8(+) T-cell responses exclusively in myeloma patients, which is indicative of pathophysiological relevance. Furthermore, in vitro priming experiments revealed that peptide-specific T-cell responses can be induced in response-naive myeloma patients. Together, our results serve to guide antigen selection for T-cell-based immunotherapy of MM.

    View details for DOI 10.1182/blood-2015-04-640532

    View details for Web of Science ID 000360540100012

    View details for PubMedID 26138685

    View details for PubMedCentralID PMC4608392