1
|
Kränke T, Tripolt-Droschl K, Röd L, Hofmann-Wellenhof R, Koppitz M, Tripolt M. New AI-algorithms on smartphones to detect skin cancer in a clinical setting-A validation study. PLoS One 2023; 18:e0280670. [PMID: 36791068 PMCID: PMC9931135 DOI: 10.1371/journal.pone.0280670] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 01/05/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The incidence of skin cancer is rising worldwide and there is medical need to optimize its early detection. This study was conducted to determine the diagnostic and risk-assessment accuracy of two new diagnosis-based neural networks (analyze and detect), which comply with the CE-criteria, in evaluating the malignant potential of various skin lesions on a smartphone. Of note, the intention of our study was to evaluate the performance of these medical products in a clinical setting for the first time. METHODS This was a prospective, single-center clinical study at one tertiary referral center in Graz, Austria. Patients, who were either scheduled for preventive skin examination or removal of at least one skin lesion were eligible for participation. Patients were assessed by at least two dermatologists and by the integrated algorithms on different mobile phones. The lesions to be recorded were randomly selected by the dermatologists. The diagnosis of the algorithm was stated as correct if it matched the diagnosis of the two dermatologists or the histology (if available). The histology was the reference standard, however, if both clinicians considered a lesion as being benign no histology was performed and the dermatologists were stated as reference standard. RESULTS A total of 238 patients with 1171 lesions (86 female; 36.13%) with an average age of 66.19 (SD = 17.05) was included. Sensitivity and specificity of the detect algorithm were 96.4% (CI 93.94-98.85) and 94.85% (CI 92.46-97.23); for the analyze algorithm a sensitivity of 95.35% (CI 93.45-97.25) and a specificity of 90.32% (CI 88.1-92.54) were achieved. DISCUSSION The studied neural networks succeeded analyzing the risk of skin lesions with a high diagnostic accuracy showing that they are sufficient tools in calculating the probability of a skin lesion being malignant. In conjunction with the wide spread use of smartphones this new AI approach opens the opportunity for a higher early detection rate of skin cancer with consecutive lower epidemiological burden of metastatic cancer and reducing health care costs. This neural network moreover facilitates the empowerment of patients, especially in regions with a low density of medical doctors. REGISTRATION Approved and registered at the ethics committee of the Medical University of Graz, Austria (Approval number: 30-199 ex 17/18).
Collapse
Affiliation(s)
- Teresa Kränke
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
- * E-mail:
| | | | - Lukas Röd
- Medical University of Graz, Graz, Austria
| | | | | | - Michael Tripolt
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
| |
Collapse
|
2
|
Noordam L, de Beijer MT, Mancham S, Vogler I, Boor PP, de Ruiter V, Luijten R, IJzermans JN, Sahin U, Bruno MJ, Sprengers D, Buschow SI, Kwekkeboom J. Systemic T-cell and humoral responses against cancer testis antigens in hepatocellular carcinoma patients. Oncoimmunology 2022; 11:2131096. [PMID: 36211805 PMCID: PMC9542711 DOI: 10.1080/2162402x.2022.2131096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related deaths worldwide due to high recurrence rates after curative treatment and being frequently diagnosed at an advanced stage. Immune-checkpoint inhibition (ICPI) has yielded impressive clinical successes in a variety of solid cancers, however results in treatment of HCC have been modest. Vaccination could be a promising treatment to synergize with ICPI and enhance response rates. Cancer testis antigens (CTAs) were recently discovered to be widely expressed in HCC and expression in macroscopically tumor-free tissues correlated with recurrence, implying the presence of micro-satellites. To determine whether CTAs are immunogenic in HCC patients, we analyzed systemic T-cell and humoral responses against seven CTAs in 38 HCC patients using a multitude of techniques; flowcytometry, ELISA and whole antigen and peptide stimulation assays. CTA-specific T-cells were detected in all (25/25) analyzed patients, of which most had a memory phenotype but did not exhibit unequivocal signs of chronic stimulation or recent antigen encounter. Proliferative CD4+ and CD8+ T-cell responses against these CTAs were found in 14/16 analyzed HCC patients. CTA-peptide stimulation-induced granzyme B, IL2, and TNFa in 8/8 analyzed patients, including two MAGEA1 peptides included based on in silico prediction. Finally, IgG responses were observed in 13/32 patients, albeit with low titers. The presence of CD4+ and CD8+ T-cells and IgG responses shows the immunogenicity of these CTAs in HCC-patients. We hypothesize that vaccines based on these tumor-specific antigens may boost preexisting CTA-specific immunity and could enhance therapeutic efficacy of ICPI in advanced HCC.
Collapse
Affiliation(s)
- Lisanne Noordam
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Monique T.A. de Beijer
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Shanta Mancham
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | | | - Patrick P.C. Boor
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Valeska de Ruiter
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Robbie Luijten
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Jan N.M. IJzermans
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | | | - Marco J. Bruno
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Sonja I. Buschow
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Jaap Kwekkeboom
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
3
|
Minott JA, van Vloten JP, Yates JGE, Chan L, Wood GA, Viloria-Petit AM, Karimi K, Petrik JJ, Wootton SK, Bridle BW. Multiplex flow cytometry-based assay for quantifying tumor- and virus-associated antibodies induced by immunotherapies. Front Immunol 2022; 13:1038340. [PMID: 36466867 PMCID: PMC9708883 DOI: 10.3389/fimmu.2022.1038340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/27/2022] [Indexed: 03/22/2024] Open
Abstract
Novel immunotherapies continue to be developed and tested for application against a plethora of diseases. The clinical translation of immunotherapies requires an understanding of their mechanisms. The contributions of antibodies in driving long-term responses following immunotherapies continue to be revealed given their diverse effector functions. Developing an in-depth understanding of the role of antibodies in treatment efficacy is required to optimize immunotherapies and improve the chance of successfully translating them into the clinic. However, analyses of antibody responses can be challenging in the context of antigen-agnostic immunotherapies, particularly in the context of cancers that lack pre-defined target antigens. As such, robust methods are needed to evaluate the capacity of a given immunotherapy to induce beneficial antibody responses, and to identify any therapy-limiting antibodies. We previously developed a comprehensive method for detecting antibody responses induced by antigen-agnostic immunotherapies for application in pre-clinical models of vaccinology and cancer therapy. Here, we extend this method to a high-throughput, flow cytometry-based assay able to identify and quantify isotype-specific virus- and tumor-associated antibody responses induced by immunotherapies using small sample volumes with rapid speed and high sensitivity. This method provides a valuable and flexible protocol for investigating antibody responses induced by immunotherapies, which researchers can use to expand their analyses and optimize their own treatment regimens.
Collapse
Affiliation(s)
- Jessica A. Minott
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | | | - Jacob G. E. Yates
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - Lily Chan
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - Geoffrey A. Wood
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | | | - Khalil Karimi
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - James J. Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Sarah K. Wootton
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - Byram W. Bridle
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
- ImmunoCeutica Inc., Cambridge, ON, Canada
| |
Collapse
|
4
|
Rodgers CB, Mustard CJ, McLean RT, Hutchison S, Pritchard AL. A B-cell or a key player? The different roles of B-cells and antibodies in melanoma. Pigment Cell Melanoma Res 2022; 35:303-319. [PMID: 35218154 PMCID: PMC9314792 DOI: 10.1111/pcmr.13031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 02/01/2022] [Accepted: 02/21/2022] [Indexed: 12/17/2022]
Abstract
The B‐cell system plays an important role in the melanoma immune response; however, consensus has yet to be reached in many facets. Here, we comprehensively review human studies only, due to fundamental differences in the humoral response with animal models. Tumour‐infiltrating B‐cells are associated with contradictory prognostic values, reflecting a lack of agreement between studies on cell subset classification and differences in the markers used, particularly the common use of a single marker not differentiating multiple subsets. Tertiary lymphoid structures (TLS) organise T‐cells and B‐cells within tumours to generate a local anti‐tumour response and TLS presence associates with improved survival in response to immune checkpoint blockade, in late‐stage disease. Autoantibody production is increased in melanoma patients and has been proposed as biomarkers for diagnosis, prognosis and treatment/toxicity response; however, no consistent targets are yet identified. The function of antibodies in an anti‐tumour response is determined by its isotype and subclass; IgG4 is immune‐suppressive and robustly correlate with poor patient survival in melanoma. We conclude that the current B‐cell literature needs careful interpretation based on the methods used and that we need a consensus of markers to define B‐cells and associated lymphoid organs. Furthermore, future studies need to not only examine antibody targets, but also isotypes when considering functional roles.
Collapse
Affiliation(s)
- Chloe B Rodgers
- Genetics and Immunology Department, Division of Biomedical Research, Institute of Health Research and Innovation, University of the Highlands and Islands, Inverness, UK
| | - Colette J Mustard
- Genetics and Immunology Department, Division of Biomedical Research, Institute of Health Research and Innovation, University of the Highlands and Islands, Inverness, UK
| | - Ryan T McLean
- Genetics and Immunology Department, Division of Biomedical Research, Institute of Health Research and Innovation, University of the Highlands and Islands, Inverness, UK
| | - Sharon Hutchison
- Genetics and Immunology Department, Division of Biomedical Research, Institute of Health Research and Innovation, University of the Highlands and Islands, Inverness, UK
| | - Antonia L Pritchard
- Genetics and Immunology Department, Division of Biomedical Research, Institute of Health Research and Innovation, University of the Highlands and Islands, Inverness, UK
| |
Collapse
|
5
|
Maniyar RR, Chakraborty S, Jarboe T, Suriano R, Wallack M, Geliebter J, Tiwari RK. Interacting Genetic Lesions of Melanoma in the Tumor Microenvironment: Defining a Viable Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1350:123-143. [PMID: 34888847 DOI: 10.1007/978-3-030-83282-7_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Melanoma is the most aggressive form of skin cancer with an estimated 106,110 newly diagnosed cases in the United States of America in 2021 leading to an approximated 7180 melanoma-induced deaths. Cancer typically arises from an accumulation of somatic mutations and can be associated with mutagenic or carcinogenic exposure. A key characteristic of melanoma is the extensive somatic mutation rate of 16.8 mutations/Mb, which is largely attributed to UV exposure. Bearing the highest mutational load, many of them occur in key driver pathways, most commonly the BRAFV600E in the mitogen-activated protein kinase (MAPK) pathway. This driver mutation is targeted clinically with FDA-approved therapies using small molecule inhibitors of oncogenic BRAFV600E and MEK, which has greatly expanded therapeutic intervention following a melanoma diagnosis. Up until 2011, therapeutic options for metastatic melanoma were limited, and treatment typically fell under the spectrum of surgery, radiotherapy, and chemotherapy.Attributed to the extensive mutation rate, as well as having the highest number of neoepitopes, melanoma is deemed to be extremely immunogenic. However, despite this highly immunogenic nature, melanoma is notorious for inducing an immunosuppressive microenvironment which can be relieved by checkpoint inhibitor therapy. The two molecules currently approved clinically are ipilimumab and nivolumab, which target the molecules CTLA-4 and PD-1, respectively.A plethora of immunomodulatory molecules exist, many with redundant functions. Additionally, these molecules are expressed not only by immune cells but also by tumor cells within the tumor microenvironment. Tumor profiling of these cell surface checkpoint molecules is necessary to optimize a clinical response. The presence of immunomodulatory molecules in melanoma, using data from The Cancer Genome Atlas and validation of expression in two model systems, human melanoma tissues and patient-derived melanoma cells, revealed that the expression levels of B and T lymphocyte attenuator (BTLA), TIM1, and CD226, concurrently with the BRAFV600E mutation status, significantly dictated overall survival in melanoma patients. These molecules, along with herpesvirus entry mediator (HVEM) and CD160, two molecules that are a part of the HVEM/BTLA/CD160 axis, had a higher expression in human melanoma tissues when compared to normal skin melanocytes and have unique roles to play in T cell activation. New links are being uncovered between the expression of immunomodulatory molecules and the BRAFV600E genetic lesion in melanoma. Small molecule inhibitors of the MAPK pathway regulate the surface expression of this multifaceted molecule, making BTLA a promising target for immuno-oncology to be targeted in combination with small molecule inhibitors, potentially alleviating T regulatory cell activation and improving patient prognosis.
Collapse
Affiliation(s)
- R R Maniyar
- Human Oncology and Pathogenesis Program, Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - S Chakraborty
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - T Jarboe
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - R Suriano
- Division of Natural Sciences, College of Mount Saint Vincent, Bronx, NY, USA
| | - M Wallack
- Department Surgery, Metropolitan Hospital, New York, NY, USA
| | - J Geliebter
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - R K Tiwari
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
6
|
Gu S, Qian L, Zhang Y, Chen K, Li Y, Wang J, Wang P. Significance of intratumoral infiltration of B cells in cancer immunotherapy: From a single cell perspective. Biochim Biophys Acta Rev Cancer 2021; 1876:188632. [PMID: 34626740 DOI: 10.1016/j.bbcan.2021.188632] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/15/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
Immunotherapy for cancer has provided new treatment approaches for malignant tumors, but there are low rates of response and high rates of resistance. The most recent sequencing method which is called single-cell RNA sequencing(scRNA-seq) determines the transcriptome at the single cell level, which allows high-resolution dynamic monitoring of the tumor microenvironment (TME) during immunotherapy. As an important part of humoral immunity, tumor-infiltrated B cells have been reported to have distinct functions in anti-tumor immunity, which are characterized by their RNA transcriptome, membrane surface receptors, and immunoglobulin secretion, suggesting great immunotherapeutic effects. On the basis of the important roles of B cells in immunotherapy reported in recent publications, we discuss the tumor-infiltrated B cells' subpopulations, differentiation trajectory, and interactions with other cells in the TME in this review, hoping to illustrate its significance in potential clinical application as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Sijia Gu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ling Qian
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yalei Zhang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Kun Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ye Li
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jia Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Peng Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
7
|
Cui M, Huang J, Zhang S, Liu Q, Liao Q, Qiu X. Immunoglobulin Expression in Cancer Cells and Its Critical Roles in Tumorigenesis. Front Immunol 2021; 12:613530. [PMID: 33841396 PMCID: PMC8024581 DOI: 10.3389/fimmu.2021.613530] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 03/08/2021] [Indexed: 12/23/2022] Open
Abstract
Traditionally, immunoglobulin (Ig) was believed to be produced by only B-lineage cells. However, increasing evidence has revealed a high level of Ig expression in cancer cells, and this Ig is named cancer-derived Ig. Further studies have shown that cancer-derived Ig shares identical basic structures with B cell-derived Ig but exhibits several distinct characteristics, including restricted variable region sequences and aberrant glycosylation. In contrast to B cell-derived Ig, which functions as an antibody in the humoral immune response, cancer-derived Ig exerts profound protumorigenic effects via multiple mechanisms, including promoting the malignant behaviors of cancer cells, mediating tumor immune escape, inducing inflammation, and activating the aggregation of platelets. Importantly, cancer-derived Ig shows promising potential for application as a diagnostic and therapeutic target in cancer patients. In this review, we summarize progress in the research area of cancer-derived Ig and discuss the perspectives of applying this novel target for the management of cancer patients.
Collapse
Affiliation(s)
- Ming Cui
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jing Huang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Shenghua Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaoyan Qiu
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
8
|
Tsuji T, Eng KH, Matsuzaki J, Battaglia S, Szender JB, Miliotto A, Gnjatic S, Bshara W, Morrison CD, Lele S, Emerson RO, Wang J, Liu S, Robins H, Lugade AA, Odunsi K. Clonality and antigen-specific responses shape the prognostic effects of tumor-infiltrating T cells in ovarian cancer. Oncotarget 2020; 11:2669-2683. [PMID: 32676168 PMCID: PMC7343634 DOI: 10.18632/oncotarget.27666] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/15/2020] [Indexed: 12/17/2022] Open
Abstract
CD8+ tumor-infiltrating lymphocytes (TILs) are not all specific for tumor antigens, but can include bystander TILs that are specific for cancer-irrelevant epitopes, and it is unknown whether the T-cell repertoire affects prognosis. To delineate the complexity of anti-tumor T-cell responses, we utilized a computational method for de novo assembly of sequences from CDR3 regions of 369 high-grade serous ovarian cancers from TCGA, and then applied deep TCR-sequencing for analyses of paired tumor and peripheral blood specimens from an independent cohort of 99 ovarian cancer patients. Strongly monoclonal T-cell repertoires were associated with favorable prognosis (PFS, HR = 0.65, 0.50-0.84, p = 0.003; OS, HR = 0.61, 0.44-0.83, p = 0.006) in TCGA cohort. In the validation cohort, we discovered that patients with low T-cell infiltration but low diversity or focused repertoires had clinical outcomes almost indistinguishable from highly-infiltrated tumors (median 21.0 months versus 15.9 months, log-rank p = 0.945). We also found that the degree of divergence of the peripheral repertoire from the TIL repertoire, and the presence of detectable spontaneous anti-tumor immune responses are important determinants of clinical outcome. We conclude that the prognostic significance of TILs in ovarian cancer is dictated by T-cell clonality, degree of overlap with peripheral repertoire, and the presence of detectable spontaneous anti-tumor immune response in the patients. These immunological phenotypes defined by the TCR repertoire may provide useful insights for identifying "TIL-low" ovarian cancer patients that may respond to immunotherapy.
Collapse
Affiliation(s)
- Takemasa Tsuji
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- These authors contributed equally to this work
| | - Kevin H Eng
- Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- These authors contributed equally to this work
| | - Junko Matsuzaki
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sebastiano Battaglia
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - J Brian Szender
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Anthony Miliotto
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sacha Gnjatic
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wiam Bshara
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Carl D Morrison
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Shashikant Lele
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Jianmin Wang
- Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Song Liu
- Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Amit A Lugade
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kunle Odunsi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
9
|
Li X, Larsson P, Ljuslinder I, Öhlund D, Myte R, Löfgren-Burström A, Zingmark C, Ling A, Edin S, Palmqvist R. Ex Vivo Organoid Cultures Reveal the Importance of the Tumor Microenvironment for Maintenance of Colorectal Cancer Stem Cells. Cancers (Basel) 2020; 12:E923. [PMID: 32290033 PMCID: PMC7226030 DOI: 10.3390/cancers12040923] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease, with varying clinical presentations and patient prognosis. Different molecular subgroups of CRC should be treated differently and therefore, must be better characterized. Organoid culture has recently been suggested as a good model to reflect the heterogeneous nature of CRC. However, organoid cultures cannot be established from all CRC tumors. The study examines which CRC tumors are more likely to generate organoids and thus benefit from ex vivo organoid drug testing. Long-term organoid cultures from 22 out of 40 CRC tumor specimens were established. It was found that organoid cultures were more difficult to establish from tumors characterized as microsatellite instable (MSI), BRAF-mutated, poorly differentiated and/or of a mucinous type. This suggests that patients with such tumors are less likely to benefit from ex vivo organoid drug testing, but it may also suggest biological difference in tumor growth. RNA sequencing analysis of tumor sections revealed that the in vivo maintenance of these non-organoid-forming tumors depends on factors related to inflammation and pathogen exposure. Furthermore, using TCGA data we could show a trend towards a worse prognosis for patients with organoid-forming tumors, suggesting also clinical differences. Results suggest that organoids are more difficult to establish from tumors characterized as MSI, BRAF-mutated, poorly differentiated and/or of a mucinous type. We further suggest that the maintenance of cell growth of these tumors in vivo may be promoted by immune-related factors and other stromal components within the tumor microenvironment.
Collapse
Affiliation(s)
- Xingru Li
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Pär Larsson
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Ingrid Ljuslinder
- Department of Radiation Sciences, Oncology, Umeå University, 90185 Umeå, Sweden; (I.L.); (D.O.); (R.M.)
| | - Daniel Öhlund
- Department of Radiation Sciences, Oncology, Umeå University, 90185 Umeå, Sweden; (I.L.); (D.O.); (R.M.)
- Wallenberg Center for Molecular Medicine, Umeå University, 90185 Umeå, Sweden
| | - Robin Myte
- Department of Radiation Sciences, Oncology, Umeå University, 90185 Umeå, Sweden; (I.L.); (D.O.); (R.M.)
| | - Anna Löfgren-Burström
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Carl Zingmark
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Agnes Ling
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Sofia Edin
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Richard Palmqvist
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| |
Collapse
|
10
|
Gangkofner DS, Holzinger D, Schroeder L, Eichmüller SB, Zörnig I, Jäger D, Wichmann G, Dietz A, Broglie MA, Herold-Mende C, Dyckhoff G, Boscolo-Rizzo P, Ezic J, Marienfeld RB, Möller P, Völkel G, Kraus JM, Kestler HA, Brunner C, Schuler PJ, Wigand M, Theodoraki MN, Doescher J, Hoffmann TK, Pawlita M, Butt J, Waterboer T, Laban S. Patterns of antibody responses to nonviral cancer antigens in head and neck squamous cell carcinoma patients differ by human papillomavirus status. Int J Cancer 2019; 145:3436-3444. [PMID: 31407331 DOI: 10.1002/ijc.32623] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/20/2019] [Accepted: 07/22/2019] [Indexed: 12/24/2022]
Abstract
There have been hints that nonviral cancer antigens are differentially expressed in human papillomavirus (HPV)-positive and HPV-negative head and neck squamous cell carcinoma (HNSCC). Antibody responses (AR) to cancer antigens may be used to indirectly determine cancer antigen expression in the tumor using a noninvasive and tissue-saving liquid biopsy. Here, we set out to characterize AR to a panel of nonviral cancer antigens in HPV-positive and HPV-negative HNSCC patients. A fluorescent microbead multiplex serology to 29 cancer antigens (16 cancer-testis antigens, 5 cancer-retina antigens and 8 oncogenes) and 29 HPV-antigens was performed in 382 HNSCC patients from five independent cohorts (153 HPV-positive and 209 HPV-negative). AR to any of the cancer antigens were found in 272/382 patients (72%). The ten most frequent AR were CT47, cTAGE5a, c-myc, LAGE-1, MAGE-A1, -A3, -A4, NY-ESO-1, SpanX-a1 and p53. AR to MAGE-A3, MAGE-A9 and p53 were found at significantly different prevalences by HPV status. An analysis of AR mean fluorescent intensity values uncovered remarkably different AR clusters by HPV status. To identify optimal antigen selections covering a maximum of patients with ≤10 AR, multiobjective optimization revealed distinct antigen selections by HPV status. We identified that AR to nonviral antigens differ by HPV status indicating differential antigen expression. Multiplex serology may be used to characterize antigen expression using serum or plasma as a tissue-sparing liquid biopsy. Cancer antigen panels should address the distinct antigen repertoire of HPV-positive and HPV-negative HNSCC.
Collapse
Affiliation(s)
- Dominik S Gangkofner
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Dana Holzinger
- Infections and Cancer Epidemiology (F022), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lea Schroeder
- Infections and Cancer Epidemiology (F022), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan B Eichmüller
- Research Group GMP & T Cell Therapy (D210), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Inka Zörnig
- National Center for Tumor Diseases (NCT) and Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Applied Tumor Immunity (D120), Heidelberg, Germany
| | - Dirk Jäger
- National Center for Tumor Diseases (NCT) and Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Applied Tumor Immunity (D120), Heidelberg, Germany
| | - Gunnar Wichmann
- Department of Otorhinolaryngology, University Hospital Leipzig, Leipzig, Germany
| | - Andreas Dietz
- Department of Otorhinolaryngology, University Hospital Leipzig, Leipzig, Germany
| | - Martina A Broglie
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Christel Herold-Mende
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Department of Neurosurgery, Division of Experimental Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Gerhard Dyckhoff
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Paolo Boscolo-Rizzo
- Department of Neurosciences, ENT Clinic and Regional Center for Head and Neck Cancer, University of Padua, Treviso, Italy
| | - Jasmin Ezic
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | | | - Peter Möller
- Institute of Pathology, University Medical Center Ulm, Ulm, Germany
| | - Gunnar Völkel
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Johann M Kraus
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Cornelia Brunner
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Patrick J Schuler
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Marlene Wigand
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Marie N Theodoraki
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Johannes Doescher
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Thomas K Hoffmann
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Michael Pawlita
- Infections and Cancer Epidemiology (F022), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julia Butt
- Infections and Cancer Epidemiology (F022), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Waterboer
- Infections and Cancer Epidemiology (F022), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Simon Laban
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| |
Collapse
|
11
|
Antibody Responses to Cancer Antigens Identify Patients with a Poor Prognosis among HPV-Positive and HPV-Negative Head and Neck Squamous Cell Carcinoma Patients. Clin Cancer Res 2019; 25:7405-7412. [DOI: 10.1158/1078-0432.ccr-19-1490] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/27/2019] [Accepted: 08/20/2019] [Indexed: 12/24/2022]
|
12
|
Garaud S, Zayakin P, Buisseret L, Rulle U, Silina K, de Wind A, Van den Eyden G, Larsimont D, Willard-Gallo K, Linē A. Antigen Specificity and Clinical Significance of IgG and IgA Autoantibodies Produced in situ by Tumor-Infiltrating B Cells in Breast Cancer. Front Immunol 2018; 9:2660. [PMID: 30515157 PMCID: PMC6255822 DOI: 10.3389/fimmu.2018.02660] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 10/29/2018] [Indexed: 12/16/2022] Open
Abstract
An important role for tumor infiltrating B lymphocytes (TIL-B) in the immune response to cancer is emerging; however, very little is known about the antigen specificity of antibodies produced in situ. The presence of IgA antibodies in the tumor microenvironment has been noted although their biological functions and clinical significance are unknown. This study used a 91-antigen microarray to examine the IgG and IgA autoantibody repertoires in breast cancer (BC). Tumor and adjacent breast tissue supernatants and plasma from BC patients together with normal breast tissue supernatants and plasma from healthy controls (patients undergoing mammary reduction and healthy blood donors) were analyzed to investigate relationships between autoantibodies and the clinical, histological and immunological features of tumors. Our data show that >84% of the BC samples tested contain autoantibodies to one or more antigens on the array, with ANKRD30BL, COPS4, and CTAG1B being most frequently reactive. Ex vivo TIL-B responses were uncoupled from systemic humoral responses in the majority of cases. A comparison of autoantibody frequencies in supernatants and plasma from patients and controls identified eight antigens that elicit BC-associated autoantibody responses. The overall prevalence of IgG and IgA autoantibodies was similar and while IgG and IgA responses were not linked they did correlate with distinct clinical, pathological and immunological features. Higher levels of ex vivo IgG responses to BC-associated antigens were associated with shorter recurrence-free survival (RFS), HER2 overexpression and lower tumor-infiltrating CD8+ T cell counts. Higher IgA levels were associated with estrogen and progesterone receptor-negative cancers but were not significantly associated with RFS. Furthermore, ex vivo IgA but not IgG autoantibodies reactive to BC-associated antigens were linked with germinal center and early memory B cell maturation and the presence of tertiary lymphoid structures suggesting that these TIL-B are activated in the tumor microenvironment. Overall, our results extend the current understanding of the antigen specificity, the biological and the clinical significance of IgG and IgA autoantibodies produced by BC TIL-B in situ.
Collapse
Affiliation(s)
- Soizic Garaud
- Molecular Immunology Unit, Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
| | - Pawel Zayakin
- Cancer Biomarker and Immunotherapy Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Laurence Buisseret
- Molecular Immunology Unit, Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
| | - Undine Rulle
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Karina Silina
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Alexandre de Wind
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Gert Van den Eyden
- Translational Cancer Research Unit Antwerp, Oncology Centre, General Hospital Sint Augustinus, Wilrijk, Belgium
| | - Denis Larsimont
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Karen Willard-Gallo
- Molecular Immunology Unit, Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
| | - Aija Linē
- Cancer Biomarker and Immunotherapy Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
- Faculty of Biology, University of Latvia, Riga, Latvia
| |
Collapse
|
13
|
B cells and antibody production in melanoma. Mamm Genome 2018; 29:790-805. [DOI: 10.1007/s00335-018-9778-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/24/2018] [Indexed: 01/12/2023]
|
14
|
A diagnostic autoantibody signature for primary cutaneous melanoma. Oncotarget 2018; 9:30539-30551. [PMID: 30093967 PMCID: PMC6078131 DOI: 10.18632/oncotarget.25669] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 06/04/2018] [Indexed: 01/16/2023] Open
Abstract
Melanoma is an aggressive form of skin cancer that is curable by surgical excision in the majority of cases, if detected at an early stage. To improve early stage melanoma detection, the development of a highly sensitive diagnostic test is of utmost importance. Here we aimed to identify antibodies to a panel of tumour associated antigens that can differentiate primary melanoma patients and healthy individuals. A total of 245 sera from primary melanoma patients and healthy volunteers were screened against a high-throughput microarray platform containing 1627 functional proteins. Following rigorous statistical analysis, we identified a combination of 10 autoantibody biomarkers that, as a panel, displays a sensitivity of 79%, specificity of 84% and an AUC of 0.828 for primary melanoma detection. This melanoma autoantibody signature may prove valuable for the development of a diagnostic blood test for routine population screening that, when used in conjunction with current melanoma diagnostic techniques, could improve the early diagnosis of this malignancy and ultimately decrease the mortality rate of patients.
Collapse
|
15
|
Michels J, Becker N, Suciu S, Kaiser I, Benner A, Kosaloglu-Yalcin Z, Agoussi S, Halama N, Pawlita M, Waterboer T, Eichmüller SB, Jäger D, Eggermont AMM, Zörnig I. Multiplex bead-based measurement of humoral immune responses against tumor-associated antigens in stage II melanoma patients of the EORTC18961 trial. Oncoimmunology 2018; 7:e1428157. [PMID: 29872552 PMCID: PMC5980408 DOI: 10.1080/2162402x.2018.1428157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/07/2018] [Accepted: 01/09/2018] [Indexed: 12/25/2022] Open
Abstract
Purpose: Determine the prognostic and predictive significance of tumor associated antigen (TAA)-specific serum antibodies in melanoma patients of a large adjuvant vaccination phase III trial. Patients and methods: Serum IgG antibodies were measured against a panel of 43 antigens by a bead-based multiplex assay in 970 stage II melanoma patients of the EORTC18961 trial, evaluating adjuvant ganglioside GM2-KLH/QS-21 vaccination versus observation. Primary end point was relapse-free survival (RFS). Patients' sera at baseline, after 12 and 48 weeks of study treatment and at the last available time point (at recurrence/remission) were evaluated. Results: Prognostic clinical variables are gender, surgical confirmation of lymph node-negative status, Breslow thickness and ulceration of the primary. Prognostic spontaneous antibody responses were associated with a significant dismal (GM2, Rhod_E2, SSX2) or good prognosis (CyclinB1, SCYE1v1) for RFS, distant metastasis-free (DMFS) or overall survival (OS). Predictive spontaneous antibody responses based on significant interaction with treatment were RhodN p = 0.02, Rab38 p = 0.04 for RFS, RhodE2 p = 0.006, Recoverin p = 0.04 for DMFS and RhodE2 p = 0.003; Recoverin p = 0.04, NA17.A p = 0.04, for OS respectively. The subgroups of patients according to antibody responses for RFS were determined for RhodN sero-negative (n = 849, HR = 1.07, p = 0.6); RhodN sero-positive (n = 121,HR = 0.42, p = 0.01) and Rab38 sero-negative (n = 682, HR = 1.12, p = 0.42), Rab38 sero-positive (n = 288, HR = 0.65, p = 0.04) patients respectively. Conclusion: We identified prognostic serum antibody responses against TAA in stage II melanoma patients. A set of antibody responses correlated with a beneficial outcome for GM2 vaccination.
Collapse
Affiliation(s)
- Judith Michels
- Department of Medical Oncology, Gustave Roussy Comprehensive Cancer Center, Villejuif/Paris-Sud, France.,Université Paris-Sud, Kremlin Bicêtre, Paris, France
| | - Natalia Becker
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Suciu
- European Organisation for Research and Treatment of Cancer, Brussels, Belgium
| | - Iris Kaiser
- Department of Medical Oncology, National Center for Tumor Diseases, Internal Medicine VI, Heidelberg University Hospital, Heidelberg, Germany
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Zeynep Kosaloglu-Yalcin
- Clincial Cooperation Unit "Applied Tumor Immunity", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sandrine Agoussi
- INSERM U981, Gustave Roussy Comprehensive Cancer Center, Villejuif/Paris-Sud, France
| | - Niels Halama
- Department of Medical Oncology, National Center for Tumor Diseases, Internal Medicine VI, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Pawlita
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Waterboer
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan B Eichmüller
- GMP & T Cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases, Internal Medicine VI, Heidelberg University Hospital, Heidelberg, Germany.,Clincial Cooperation Unit "Applied Tumor Immunity", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander M M Eggermont
- Department of Medical Oncology, Gustave Roussy Comprehensive Cancer Center, Villejuif/Paris-Sud, France.,Université Paris-Sud, Kremlin Bicêtre, Paris, France
| | - Inka Zörnig
- Department of Medical Oncology, National Center for Tumor Diseases, Internal Medicine VI, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
16
|
Haag GM, Zoernig I, Hassel JC, Halama N, Dick J, Lang N, Podola L, Funk J, Ziegelmeier C, Juenger S, Bucur M, Umansky L, Falk CS, Freitag A, Karapanagiotou-Schenkel I, Beckhove P, Enk A, Jaeger D. Phase II trial of ipilimumab in melanoma patients with preexisting humoural immune response to NY-ESO-1. Eur J Cancer 2018; 90:122-129. [PMID: 29306769 DOI: 10.1016/j.ejca.2017.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 12/07/2017] [Indexed: 11/20/2022]
Abstract
BACKGROUND Immune checkpoint therapy has dramatically changed treatment options in patients with metastatic melanoma. However, a relevant part of patients still does not respond to treatment. Data regarding the prognostic or predictive significance of preexisting immune responses against tumour antigens are conflicting. Retrospective data suggested a higher clinical benefit of ipilimumab in melanoma patients with preexisting NY-ESO-1-specific immunity. PATIENTS AND METHODS Twenty-five patients with previously untreated or treated metastatic melanoma and preexisting humoural immune response against NY-ESO-1 received ipilimumab at a dose of 10 mg/kg in week 1, 4, 7, 10 followed by 3-month maintenance treatment for a maximum of 48 weeks. Primary endpoint was the disease control rate (irCR, irPR or irSD) according to immune-related response criteria (irRC). Secondary endpoints included the disease control rate according to RECIST criteria, progression-free survival and overall survival (OS). Humoural and cellular immune responses against NY-ESO-1 were analysed from blood samples. RESULTS Disease control rate according to irRC was 52%, irPR was observed in 36% of patients. Progression-free survival according to irRC was 7.8 months, according to RECIST criteria it was 2.9 months. Median OS was 22.7 months; the corresponding 1-year survival rate was 66.8%. Treatment-related grade 3 AEs occurred in 36% with no grade 4-5 AEs. No clear association was found between the presence of NY-ESO-1-specific cellular or humoural immune responses and clinical activity. CONCLUSION Ipilimumab demonstrated clinically relevant activity within this biomarker-defined population. NY-ESO-1 positivity, as a surrogate for a preexisting immune response against tumour antigens, might help identifying patients with a superior outcome from immune checkpoint blockade. CLINICAL TRIAL INFORMATION NCT01216696.
Collapse
Affiliation(s)
- G M Haag
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Germany.
| | - I Zoernig
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - J C Hassel
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - N Halama
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - J Dick
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - N Lang
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - L Podola
- Translational Immunology, National Center for Tumor Diseases, Heidelberg, Germany
| | - J Funk
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - C Ziegelmeier
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - S Juenger
- Translational Immunology, National Center for Tumor Diseases, Heidelberg, Germany
| | - M Bucur
- Translational Immunology, National Center for Tumor Diseases, Heidelberg, Germany
| | - L Umansky
- Translational Immunology, National Center for Tumor Diseases, Heidelberg, Germany
| | - C S Falk
- Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany
| | - A Freitag
- NCT Trial Center, National Center for Tumor Diseases, Heidelberg, Germany
| | | | - P Beckhove
- Translational Immunology, National Center for Tumor Diseases, Heidelberg, Germany; Regensburg Center for Interventional Immunology, University Hospital Regensburg, Germany
| | - A Enk
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - D Jaeger
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Germany; Clinical Cooperation Unit "Applied Tumor-Immunity", German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
17
|
Guy TV, Terry AM, Bolton HA, Hancock DG, Zhu E, Brink R, McGuire HM, Shklovskaya E, Fazekas de St. Groth B. Collaboration between tumor-specific CD4+ T cells and B cells in anti-cancer immunity. Oncotarget 2017; 7:30211-29. [PMID: 27121060 PMCID: PMC5058675 DOI: 10.18632/oncotarget.8797] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/06/2016] [Indexed: 12/02/2022] Open
Abstract
The role of B cells and antibodies in anti-tumor immunity is controversial, with both positive and negative effects reported in animal models and clinical studies. We developed a murine B16.F10 melanoma model to study the effects of collaboration between tumor-specific CD4+ T cells and B cells on tumor control. By incorporating T cell receptor transgenic T cells and B cell receptor isotype switching B cells, we were able to track the responses of tumor-reactive T and B cells and the development of anti-tumor antibodies in vivo. In the presence of tumor-specific B cells, the number of tumor-reactive CD4+ T cells was reduced in lymphoid tissues and the tumor itself, and this correlated with poor tumor control. B cells had little effect on the Th1 bias of the CD4+ T cell response, and the number of induced FoxP3+ regulatory cells (iTregs) generated from within the original naive CD4+ T cell inoculum was unrelated to the degree of B cell expansion. In response to CD4+ T cell help, B cells produced a range of isotype-switched anti-tumor antibodies, principally IgG1, IgG2a/c and IgG2b. In the absence of CD4+ T cells, B cells responded to agonistic anti-CD40 administration by switching to production of IgG2a/c and, to a lesser extent, IgG1, IgG3, IgA and IgE, which reduced the number of lung metastases after i.v. tumor inoculation but had no effect on the growth of subcutaneous tumors.
Collapse
Affiliation(s)
- Thomas V Guy
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Alexandra M Terry
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Holly A Bolton
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - David G Hancock
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Erhua Zhu
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Robert Brink
- B Cell Laboratory, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Helen M McGuire
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Elena Shklovskaya
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Barbara Fazekas de St. Groth
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
18
|
Chen H, Werner S, Butt J, Zörnig I, Knebel P, Michel A, Eichmüller SB, Jäger D, Waterboer T, Pawlita M, Brenner H. Prospective evaluation of 64 serum autoantibodies as biomarkers for early detection of colorectal cancer in a true screening setting. Oncotarget 2017; 7:16420-32. [PMID: 26909861 PMCID: PMC4941325 DOI: 10.18632/oncotarget.7500] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/05/2016] [Indexed: 02/07/2023] Open
Abstract
Novel blood-based screening tests are strongly desirable for early detection of colorectal cancer (CRC). We aimed to identify and evaluate autoantibodies against tumor-associated antigens as biomarkers for early detection of CRC. 380 clinically identified CRC patients and samples of participants with selected findings from a cohort of screening colonoscopy participants in 2005–2013 (N=6826) were included in this analysis. Sixty-four serum autoantibody markers were measured by multiplex bead-based serological assays. A two-step approach with selection of biomarkers in a training set, and validation of findings in a validation set, the latter exclusively including participants from the screening setting, was applied. Anti-MAGEA4 exhibited the highest sensitivity for detecting early stage CRC and advanced adenoma. Multi-marker combinations substantially increased sensitivity at the price of a moderate loss of specificity. Anti-TP53, anti-IMPDH2, anti-MDM2 and anti-MAGEA4 were consistently included in the best-performing 4-, 5-, and 6-marker combinations. This four-marker panel yielded a sensitivity of 26% (95% CI, 13–45%) for early stage CRC at a specificity of 90% (95% CI, 83–94%) in the validation set. Notably, it also detected 20% (95% CI, 13–29%) of advanced adenomas. Taken together, the identified biomarkers could contribute to the development of a useful multi-marker blood-based test for CRC early detection.
Collapse
Affiliation(s)
- Hongda Chen
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Simone Werner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julia Butt
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Inka Zörnig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Internal Medicine VI, University of Heidelberg, Heidelberg, Germany
| | - Phillip Knebel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Angelika Michel
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan B Eichmüller
- GMP & T cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Internal Medicine VI, University of Heidelberg, Heidelberg, Germany
| | - Tim Waterboer
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Pawlita
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
19
|
Chen H, Qian J, Werner S, Cuk K, Knebel P, Brenner H. Development and validation of a panel of five proteins as blood biomarkers for early detection of colorectal cancer. Clin Epidemiol 2017; 9:517-526. [PMID: 29184444 PMCID: PMC5672848 DOI: 10.2147/clep.s144171] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objective Reliable noninvasive biomarkers for early detection of colorectal cancer (CRC) are highly desirable for efficient population-based screening with high adherence rates. We aimed to discover and validate blood-based protein markers for the early detection of CRC. Patients and methods A two-stage design with a discovery and a validation set was used. In the discovery phase, plasma levels of 92 protein markers and serum levels of TP53 autoantibody were measured in 226 clinically recruited CRC patients and 118 controls who were free of colorectal neoplasms at screening colonoscopy. An algorithm predicting the presence of CRC was derived by Lasso regression and validated in a validation set consisting of all available 41 patients with CRC and a representative sample of 106 participants with advanced adenomas and 107 controls free of neoplasm from a large screening colonoscopy cohort (N=6018). Receiver operating characteristic (ROC) analyses were conducted to evaluate the diagnostic performance of individual biomarkers and biomarker combinations. Results An algorithm based on growth differentiation factor 15 (GDF-15), amphiregulin (AREG), Fas antigen ligand (FasL), Fms-related tyrosine kinase 3 ligand (Flt3L) and TP53 autoantibody was constructed. In the validation set, the areas under the curves of this five-marker algorithm were 0.82 (95% CI, 0.74–0.90) for detecting CRC and 0.60 (95% CI, 0.52–0.69) for detecting advanced adenomas. At cutoffs yielding 90% specificity, the sensitivities (95% CI) for detecting CRC and advanced adenomas were 56.4% (38.4%–71.8%) and 22.0% (13.4%–35.4%), respectively. The five-marker panel showed similar diagnostic efficacy for the detection of early- and late-stage CRC. Conclusion The identified most promising biomarkers could contribute to the development of powerful blood-based tests for CRC screening in the future.
Collapse
Affiliation(s)
- Hongda Chen
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Program Office for Cancer Screening in Urban China, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Jing Qian
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Simone Werner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katarina Cuk
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Phillip Knebel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
20
|
Guy TV, Terry AM, Bolton HA, Hancock DG, Shklovskaya E, Fazekas de St. Groth B. Pro- and anti-tumour effects of B cells and antibodies in cancer: a comparison of clinical studies and preclinical models. Cancer Immunol Immunother 2016; 65:885-96. [PMID: 27222052 PMCID: PMC11029718 DOI: 10.1007/s00262-016-1848-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 05/12/2016] [Indexed: 12/15/2022]
Abstract
The primary immune role of B cells is to produce antibodies, but they can also influence T cell function via antigen presentation and, in some contexts, immune regulation. Whether their roles in tumour immunity are similar to those in other chronic immune responses such as autoimmunity and chronic infection, where both pro- and anti-inflammatory roles have been described, remains controversial. Many studies have aimed to define the role of B cells in antitumor immune responses, but despite this considerable body of work, it is not yet possible to predict how they will affect immunity to any given tumour. In many human cancers, the presence of tumour-infiltrating B cells and tumour-reactive antibodies correlates with extended patient survival, and this clinical observation is supported by data from some animal models. On the other hand, T cell responses can be adversely affected by B cell production of immunoregulatory cytokines, a phenomenon that has been demonstrated in humans and in animal models. The isotype and concentration of tumour-reactive antibodies may also influence tumour progression. Recruitment of B cells into tumours may directly reflect the subtype and strength of the anti-tumour T cell response. As the response becomes chronic, B cells may attenuate T cell responses in an attempt to decrease host damage, similar to their described role in chronic infection and autoimmunity. Understanding how B cell responses in cancer are related to the effectiveness of the overall anti-tumour response is likely to aid in the development of new therapeutic interventions against cancer.
Collapse
Affiliation(s)
- Thomas V Guy
- T Cell Biology Research Program, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW, 2042, Australia
- Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Alexandra M Terry
- T Cell Biology Research Program, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW, 2042, Australia
- Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Holly A Bolton
- T Cell Biology Research Program, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW, 2042, Australia
- Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - David G Hancock
- T Cell Biology Research Program, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW, 2042, Australia
- Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Elena Shklovskaya
- T Cell Biology Research Program, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW, 2042, Australia
- Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Barbara Fazekas de St. Groth
- T Cell Biology Research Program, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW, 2042, Australia.
- Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
21
|
Werner S, Chen H, Butt J, Michel A, Knebel P, Holleczek B, Zörnig I, Eichmüller SB, Jäger D, Pawlita M, Waterboer T, Brenner H. Evaluation of the diagnostic value of 64 simultaneously measured autoantibodies for early detection of gastric cancer. Sci Rep 2016; 6:25467. [PMID: 27140836 PMCID: PMC4853774 DOI: 10.1038/srep25467] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/18/2016] [Indexed: 02/08/2023] Open
Abstract
Autoantibodies against tumor-associated antigens (TAAs) have been suggested as biomarkers for early detection of gastric cancer. However, studies that systematically assess the diagnostic performance of a large number of autoantibodies are rare. Here, we used bead-based multiplex serology to simultaneously measure autoantibody responses against 64 candidate TAAs in serum samples from 329 gastric cancer patients, 321 healthy controls and 124 participants with other diseases of the upper digestive tract. At 98% specificity, sensitivities for the 64 tested autoantibodies ranged from 0–12% in the training set and a combination of autoantibodies against five TAAs (MAGEA4 + CTAG1 + TP53 + ERBB2_C + SDCCAG8) was able to detect 32% of the gastric cancer patients at a specificity of 87% in the validation set. Sensitivities for early and late stage gastric cancers were similar, while chronic atrophic gastritis, a precursor lesion of gastric cancer, was not detectable. However, the 5-marker combination also detected 26% of the esophageal cancer patients. In conclusion, the tested autoantibodies and combinations alone did not reach sufficient sensitivity for gastric cancer screening. Nevertheless, some autoantibodies, such as anti-MAGEA4, anti-CTAG1 or anti-TP53 and their combinations could possibly contribute to the development of cancer early detection tests (not necessarily restricted to gastric cancer) when being combined with other markers.
Collapse
Affiliation(s)
- Simone Werner
- Division of Clinical Epidemiology and Aging Research, DKFZ, Heidelberg, Germany
| | - Hongda Chen
- Division of Clinical Epidemiology and Aging Research, DKFZ, Heidelberg, Germany
| | - Julia Butt
- Division of Molecular Diagnostics of Oncogenic Infections, DKFZ, Heidelberg, Germany
| | - Angelika Michel
- Division of Molecular Diagnostics of Oncogenic Infections, DKFZ, Heidelberg, Germany
| | - Phillip Knebel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | | | - Inka Zörnig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) and Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan B Eichmüller
- GMP &T cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) and Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Pawlita
- Division of Molecular Diagnostics of Oncogenic Infections, DKFZ, Heidelberg, Germany
| | - Tim Waterboer
- Division of Molecular Diagnostics of Oncogenic Infections, DKFZ, Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, DKFZ, Heidelberg, Germany.,Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
22
|
Pfirschke C, Gebhardt C, Zörnig I, Pritsch M, Eichmüller SB, Jäger D, Enk A, Beckhove P. T cell responses in early-stage melanoma patients occur frequently and are not associated with humoral response. Cancer Immunol Immunother 2015; 64:1369-81. [PMID: 26160687 PMCID: PMC11028448 DOI: 10.1007/s00262-015-1739-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 06/20/2015] [Indexed: 11/26/2022]
Abstract
Endogenous tumor-specific T cells are detectable in patients with different tumor types including malignant melanoma (MM). They can control tumor growth, have impact on patient survival and correlate with improved clinical response to immune checkpoint therapy. Thus, they may represent a potent biomarker for respective treatment decisions. So far, major target antigens of endogenous MM-reactive T cells have not been determined systematically. Instead, autoantibodies are discussed as surrogate parameter for MM-specific T cells. Throughout a period of more than 60 days after tumor resection, we therefore determined in 38 non-metastasized primary MM patients and in healthy individuals by IFNγ ELISpot and bead-based fluorescent multiplex assay major target antigens of spontaneous T cell and humoral responses using a broad panel of MM antigens and assessed the presence and suppressive impact of MM-reactive regulatory T cells (Tregs). We show that MM-reactive T cells are frequent in MM patients, transiently increase after tumor removal and are mostly directed against Melan-A/MART-1, Tyrosinase, NA17-A and p53. MM-specific Tregs were only detected in few patients and inhibited MM-reactive T cells particularly early after tumor resection. Tumor-specific autoantibodies occurred in most patients, but did not correlate with T cell responses. Thus, endogenous antibodies may not be reliable surrogate parameters of MM-reactive T cells.
Collapse
Affiliation(s)
- Christina Pfirschke
- Division of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Christoffer Gebhardt
- Skin Cancer Unit, DKFZ, Heidelberg, Germany
- Department of Dermatology, Ruperto-Carola University of Heidelberg, Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruperto-Carola University of Heidelberg, Mannheim, Germany
| | - Inka Zörnig
- Department of Medical Oncology, NCT, Heidelberg University Hospital, Heidelberg, Germany
| | - Maria Pritsch
- Division of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Stefan B Eichmüller
- Division of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, NCT, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander Enk
- Department of Dermatology, Ruperto-Carola University of Heidelberg, Heidelberg, Germany
| | - Philipp Beckhove
- Division of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.
- Regensburg Center for Interventional Immunology (RCI), University of Regensburg, Regensburg, Germany.
- University Clinic of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
23
|
Reed CM, Cresce ND, Mauldin IS, Slingluff CL, Olson WC. Vaccination with Melanoma Helper Peptides Induces Antibody Responses Associated with Improved Overall Survival. Clin Cancer Res 2015; 21:3879-87. [PMID: 25967144 PMCID: PMC4558239 DOI: 10.1158/1078-0432.ccr-15-0233] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/04/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE A melanoma vaccine incorporating six peptides designed to induce helper T-cell responses to melanoma antigens has induced Th1-dominant CD4(+) T-cell responses in most patients, and induced durable clinical responses or stable disease in 24% of evaluable patients. The present study tested whether this vaccine also induced antibody (Ab) responses to each peptide, and whether Ab responses were associated with T-cell responses and with clinical outcome. EXPERIMENTAL DESIGN Serum samples were studied from 35 patients with stage III-IV melanomas vaccinated with 6 melanoma helper peptides (6MHP). IgG Ab responses were measured by ELISA. Associations with immune response and overall survival were assessed by log-rank test and χ(2) analysis of Kaplan-Meier data. RESULTS Ab responses to 6MHP were detected by week 7 in 77% of patients, and increased to peak 6 weeks after the last vaccine and persisted to 6 months. Ab responses were induced most frequently to longer peptides. Of those with T-cell responses, 82% had early Ab responses. Survival was improved for patients with early Ab response (P = 0.0011) or with early T-cell response (P < 0.006), and was best for those with both Ab and T-cell responses (P = 0.0002). CONCLUSIONS Vaccination with helper peptides induced both Ab responses and T-cell responses, associated with favorable clinical outcome. Such immune responses may predict favorable clinical outcome to guide combination immunotherapy. Further studies are warranted to understand mechanisms of interaction of these Abs, T-cell responses, and tumor control.
Collapse
Affiliation(s)
- Caroline M Reed
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Nicole D Cresce
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Ileana S Mauldin
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Craig L Slingluff
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Walter C Olson
- Department of Surgery, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
24
|
Besse B, Charrier M, Lapierre V, Dansin E, Lantz O, Planchard D, Le Chevalier T, Livartoski A, Barlesi F, Laplanche A, Ploix S, Vimond N, Peguillet I, Théry C, Lacroix L, Zoernig I, Dhodapkar K, Dhodapkar M, Viaud S, Soria JC, Reiners KS, Pogge von Strandmann E, Vély F, Rusakiewicz S, Eggermont A, Pitt JM, Zitvogel L, Chaput N. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology 2015; 5:e1071008. [PMID: 27141373 PMCID: PMC4839329 DOI: 10.1080/2162402x.2015.1071008] [Citation(s) in RCA: 539] [Impact Index Per Article: 59.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 07/03/2015] [Accepted: 07/05/2015] [Indexed: 01/11/2023] Open
Abstract
Dendritic cell-derived exosomes (Dex) are small extracellular vesicles secreted by viable dendritic cells. In the two phase-I trials that we conducted using the first generation of Dex (IFN-γ-free) in end-stage cancer, we reported that Dex exerted natural killer (NK) cell effector functions in patients. A second generation of Dex (IFN-γ-Dex) was manufactured with the aim of boosting NK and T cell immune responses. We carried out a phase II clinical trial testing the clinical benefit of IFN-γ-Dex loaded with MHC class I- and class II-restricted cancer antigens as maintenance immunotherapy after induction chemotherapy in patients bearing inoperable non-small cell lung cancer (NSCLC) without tumor progression. The primary endpoint was to observe at least 50% of patients with progression-free survival (PFS) at 4 mo after chemotherapy cessation. Twenty-two patients received IFN-γ-Dex. One patient exhibited a grade three hepatotoxicity. The median time to progression was 2.2 mo and median overall survival (OS) was 15 mo. Seven patients (32%) experienced stabilization of >4 mo. The primary endpoint was not reached. An increase in NKp30-dependent NK cell functions were evidenced in a fraction of these NSCLC patients presenting with defective NKp30 expression. Importantly, MHC class II expression levels of the final IFN-γ-Dex product correlated with expression levels of the NKp30 ligand BAG6 on Dex, and with NKp30-dependent NK functions, the latter being associated with longer progression-free survival. This phase II trial confirmed the capacity of Dex to boost the NK cell arm of antitumor immunity in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Benjamin Besse
- Gustave Roussy Cancer Campus, Villejuif, France; Département de Médecine Oncologique (Unité thorax), Gustave Roussy Cancer Campus, Villejuif, France; Faculté de médecine, Université Paris Sud, Le Kremlin-Bicêtre, France
| | - Mélinda Charrier
- Gustave Roussy Cancer Campus, Villejuif, France; Faculté de médecine, Université Paris Sud, Le Kremlin-Bicêtre, France; Laboratoire d'Immunomonitoring en Oncologie, UMS 3655 CNRS / US 23 INSERM Gustave Roussy Cancer Campus, Villejuif, France; Centre d'Investigation Clinique en Biothérapies (CICBT) 1428, Villejuif, France
| | - Valérie Lapierre
- Gustave Roussy Cancer Campus, Villejuif, France; Laboratoire de Thérapie Cellulaire, Gustave Roussy Cancer Campus, Villejuif, France
| | - Eric Dansin
- Département d'oncologie générale, CLCC Oscar Lambret , Lille, France
| | - Olivier Lantz
- Centre d'Investigation Clinique en Biothérapies (CICBT) 1428, Villejuif, France; Laboratoire d'Immunologie Clinique, Institut Curie, Paris, France; Inserm U932, Institut Curie, Paris, France
| | - David Planchard
- Département de Médecine Oncologique (Unité thorax), Gustave Roussy Cancer Campus , Villejuif, France
| | - Thierry Le Chevalier
- Département de Médecine Oncologique (Unité thorax), Gustave Roussy Cancer Campus , Villejuif, France
| | - Alain Livartoski
- Institut Curie, Département de médecine oncologique , Paris, France
| | - Fabrice Barlesi
- Service d'Oncologie Multidisciplinaire & Innovations Thérapeutiques, Université Aix Marseille, Assistance Publique Hôpitaux de Marseille , Marseille, France
| | - Agnès Laplanche
- Département de Biostatistique et d'épidémiologie, Gustave Roussy Cancer Campus , Villejuif, France
| | - Stéphanie Ploix
- Centre d'Investigation Clinique en Biothérapies (CICBT) 1428, Villejuif, France
| | - Nadège Vimond
- Laboratoire d'Immunomonitoring en Oncologie, UMS 3655 CNRS / US 23 INSERM Gustave Roussy Cancer Campus, Villejuif, France; Centre d'Investigation Clinique en Biothérapies (CICBT) 1428, Villejuif, France
| | - Isabelle Peguillet
- Centre d'Investigation Clinique en Biothérapies (CICBT) 1428, Villejuif, France; Laboratoire d'Immunologie Clinique, Institut Curie, Paris, France; Inserm U932, Institut Curie, Paris, France
| | - Clotilde Théry
- Centre d'Investigation Clinique en Biothérapies (CICBT) 1428, Villejuif, France; Inserm U932, Institut Curie, Paris, France
| | - Ludovic Lacroix
- Département de biologie et pathologie médicale, Gustave Roussy Cancer Campus, Villejuif, France; Laboratoire de Recherche Translationnelle, UMS 3655 CNRS / US 23 INSERM Gustave Roussy Cancer Campus, Villejuif, France
| | - Inka Zoernig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) and Heidelberg University Hospital , Heidelberg, Germany
| | - Kavita Dhodapkar
- Department of Pediatrics, Yale School of Medicine, Yale University, New Haven, CT, USA; Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Madhav Dhodapkar
- Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, USA; Department of Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA; Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Sophie Viaud
- Gustave Roussy Cancer Campus, Villejuif, France; Faculté de médecine, Université Paris Sud, Le Kremlin-Bicêtre, France; INSERM, U1015, Villejuif, France
| | - Jean-Charles Soria
- Gustave Roussy Cancer Campus, Villejuif, France; Faculté de médecine, Université Paris Sud, Le Kremlin-Bicêtre, France; INSERM, U981, Villejuif, France; Département d'Innovation Thérapeutique et d'Essais Précoces, Gustave Roussy Cancer Campus, Villejuif, France
| | - Katrin S Reiners
- Department of Internal Medicine I, University Hospital of Cologne , Cologne, Germany
| | | | - Frédéric Vély
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Case 906, France; INSERM, U1104, Marseille, France; CNRS, UMR7280, Marseille, France; Laboratoire d'immunologie- Hôpital de la Conception, Assistance Publique - Hôpitaux de Marseille, Marseille, France
| | - Sylvie Rusakiewicz
- Gustave Roussy Cancer Campus, Villejuif, France; Centre d'Investigation Clinique en Biothérapies (CICBT) 1428, Villejuif, France; INSERM, U1015, Villejuif, France
| | - Alexander Eggermont
- Gustave Roussy Cancer Campus, Villejuif, France; Faculté de médecine, Université Paris Sud, Le Kremlin-Bicêtre, France; INSERM, U1015, Villejuif, France
| | - Jonathan M Pitt
- Gustave Roussy Cancer Campus, Villejuif, France; Faculté de médecine, Université Paris Sud, Le Kremlin-Bicêtre, France; INSERM, U1015, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France; Faculté de médecine, Université Paris Sud, Le Kremlin-Bicêtre, France; Centre d'Investigation Clinique en Biothérapies (CICBT) 1428, Villejuif, France; INSERM, U1015, Villejuif, France
| | - Nathalie Chaput
- Gustave Roussy Cancer Campus, Villejuif, France; Laboratoire d'Immunomonitoring en Oncologie, UMS 3655 CNRS / US 23 INSERM Gustave Roussy Cancer Campus, Villejuif, France; Centre d'Investigation Clinique en Biothérapies (CICBT) 1428, Villejuif, France; Laboratoire de Thérapie Cellulaire, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|