Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 177(5): 1330-1345.e18, 2019 05 16.
Article in English | MEDLINE | ID: mdl-30982598

ABSTRACT

Breast cancer is a heterogeneous disease. Tumor cells and associated healthy cells form ecosystems that determine disease progression and response to therapy. To characterize features of breast cancer ecosystems and their associations with clinical data, we analyzed 144 human breast tumor and 50 non-tumor tissue samples using mass cytometry. The expression of 73 proteins in 26 million cells was evaluated using tumor and immune cell-centric antibody panels. Tumors displayed individuality in tumor cell composition, including phenotypic abnormalities and phenotype dominance. Relationship analyses between tumor and immune cells revealed characteristics of ecosystems related to immunosuppression and poor prognosis. High frequencies of PD-L1+ tumor-associated macrophages and exhausted T cells were found in high-grade ER+ and ER- tumors. This large-scale, single-cell atlas deepens our understanding of breast tumor ecosystems and suggests that ecosystem-based patient classification will facilitate identification of individuals for precision medicine approaches targeting the tumor and its immunoenvironment.


Subject(s)
Breast Neoplasms , Immune Tolerance , Lymphocytes, Tumor-Infiltrating , Macrophages , Tumor Microenvironment/immunology , B7-H1 Antigen/immunology , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Disease-Free Survival , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Macrophages/immunology , Macrophages/pathology , Neoplasm Proteins/immunology , Survival Rate
2.
Cell ; 169(4): 736-749.e18, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475899

ABSTRACT

Immune cells in the tumor microenvironment modulate cancer progression and are attractive therapeutic targets. Macrophages and T cells are key components of the microenvironment, yet their phenotypes and relationships in this ecosystem and to clinical outcomes are ill defined. We used mass cytometry with extensive antibody panels to perform in-depth immune profiling of samples from 73 clear cell renal cell carcinoma (ccRCC) patients and five healthy controls. In 3.5 million measured cells, we identified 17 tumor-associated macrophage phenotypes, 22 T cell phenotypes, and a distinct immune composition correlated with progression-free survival, thereby presenting an in-depth human atlas of the immune tumor microenvironment in this disease. This study revealed potential biomarkers and targets for immunotherapy development and validated tools that can be used for immune profiling of other tumor types.


Subject(s)
Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Tumor Microenvironment , Humans , Image Cytometry , Immune Tolerance , Kidney/cytology , Macrophages/immunology , Macrophages/pathology , Single-Cell Analysis , T-Lymphocytes/immunology , T-Lymphocytes/pathology
3.
Eur J Immunol ; : e2451207, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38980268

ABSTRACT

Tertiary lymphoid structures (TLS) resemble follicles of secondary lymphoid organs and develop in nonlymphoid tissues during inflammation and cancer. Which cell types and signals drive the development of TLS is largely unknown. To investigate early events of TLS development in the lungs, we repeatedly instilled p(I:C) plus ovalbumin (Ova) intranasally. This induced TLS ranging from lymphocytic aggregates to organized and functional structures containing germinal centers. We found that TLS development is independent of FAP+ fibroblasts, alveolar macrophages, or CCL19 but crucially depends on type I interferon (IFN-I). Mechanistically, IFN-I initiates two synergistic pathways that culminate in the development of TLS. On the one hand, IFN-I induces lymphotoxin (LT)α in lymphoid cells, which stimulate stromal cells to produce the B-cell-attracting chemokine CXCL13 through LTßR-signaling. On the other hand, IFN-I is sensed by stromal cells that produce the T-cell-attracting chemokines CXCL9, CXCL10 as well as CCL19 and CCL21 independently of LTßR. Consequently, B-cell aggregates develop within a week, whereas follicular dendritic cells and germinal centers appear after 3 weeks. Thus, sustained production of IFN-I together with an antigen is essential for the induction of functional TLS in the lungs.

4.
Immunity ; 42(4): 767-77, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25888260

ABSTRACT

Radiotherapy induces DNA damage and cell death, but recent data suggest that concomitant immune stimulation is an integral part of the therapeutic action of ionizing radiation. It is poorly understood how radiotherapy supports tumor-specific immunity. Here we report that radiotherapy induced tumor cell death and transiently activated complement both in murine and human tumors. The local production of pro-inflammatory anaphylatoxins C3a and C5a was crucial to the tumor response to radiotherapy and concomitant stimulation of tumor-specific immunity. Dexamethasone, a drug frequently given during radiotherapy, limited complement activation and the anti-tumor effects of the immune system. Overall, our findings indicate that anaphylatoxins are key players in radiotherapy-induced tumor-specific immunity and the ensuing clinical responses.


Subject(s)
Complement C3a/immunology , Complement C5a/immunology , Gene Expression Regulation, Neoplastic/immunology , Immunity, Innate/radiation effects , Melanoma, Experimental/immunology , Skin Neoplasms/immunology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Complement Activation , Complement C3a/genetics , Complement C5a/genetics , Dexamethasone/pharmacology , Gamma Rays , Humans , Immunity, Innate/drug effects , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Melanoma, Experimental/radiotherapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Complement/genetics , Receptors, Complement/immunology , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/radiotherapy , Tumor Burden/radiation effects
5.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Article in English | MEDLINE | ID: mdl-34183415

ABSTRACT

The liver is a major metastatic target organ, and little is known about the role of immunity in controlling hepatic metastases. Here, we discovered that the concerted and nonredundant action of two innate lymphocyte subpopulations, conventional natural killer cells (cNKs) and tissue-resident type I innate lymphoid cells (trILC1s), is essential for antimetastatic defense. Using different preclinical models for liver metastasis, we found that trILC1 controls metastatic seeding, whereas cNKs restrain outgrowth. Whereas the killing capacity of trILC1s was not affected by the metastatic microenvironment, the phenotype and function of cNK cells were affected in a cancer type-specific fashion. Thus, individual cancer cell lines orchestrate the emergence of unique cNK subsets, which respond differently to tumor-derived factors. Our findings will contribute to the development of therapies for liver metastasis involving hepatic innate cells.


Subject(s)
Immunity, Innate/immunology , Killer Cells, Natural/immunology , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Lymphocytes/immunology , Animals , Female , Gene Expression Regulation, Neoplastic , Integrin alpha1/metabolism , Interleukin-15/metabolism , Liver/immunology , Liver/pathology , Liver Neoplasms/genetics , Mice , Mice, Inbred C57BL , RNA-Seq , Single-Cell Analysis , Transcriptome/genetics , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
6.
Eur J Immunol ; 51(7): 1799-1808, 2021 07.
Article in English | MEDLINE | ID: mdl-33759186

ABSTRACT

Mononuclear phagocytes consisting of monocytes, macrophages, and DCs play a complex role in tumor development by either promoting or restricting tumor growth. Cutaneous squamous cell carcinoma (cSCC) is the second most common nonmelanoma skin cancer arising from transformed epidermal keratinocytes. While present at high numbers, the role of tumor-infiltrating and resident myeloid cells in the formation of cSCC is largely unknown. Using transgenic mice and depleting antibodies to eliminate specific myeloid cell types in the skin, we investigated the involvement of mononuclear phagocytes in the development of UV-induced cSCC in K14-HPV8-E6 transgenic mice. Although resident Langerhans cells were enriched in the tumor, their contribution to tumor formation was negligible. Equally, dermal macrophages were dispensable for the development of cSCC. In contrast, mice lacking circulating monocytes were completely resistant to UV-induced cSCC, indicating that monocytes promote tumor development. Collectively, these results demonstrate a critical role for classical monocytes in the initiation of skin cancer.


Subject(s)
Carcinogenesis/pathology , Epidermis/pathology , Monocytes/pathology , Skin Neoplasms/pathology , Ultraviolet Rays/adverse effects , Animals , Carcinogenesis/radiation effects , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Epidermis/radiation effects , Female , Keratinocytes/pathology , Keratinocytes/radiation effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/radiation effects , Skin/pathology , Skin/radiation effects
7.
Immunity ; 37(2): 264-75, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22902234

ABSTRACT

Mature dendritic cells (DCs) are established as unrivaled antigen-presenting cells (APCs) in the initiation of immune responses, whereas steady-state DCs induce peripheral T cell tolerance. Using various genetic approaches, we depleted CD11c(+) DCs in mice and induced autoimmune CNS inflammation. Unexpectedly, mice lacking DCs developed aggravated disease compared to control mice. Furthermore, when we engineered DCs to present a CNS-associated autoantigen in an induced manner, we found robust tolerance that prevented disease, which coincided with an upregulation of the PD-1 receptor on antigen-specific T cells. Additionally, we showed that PD-1 was necessary for DC-mediated induction of regulatory T cells. Our results show that a reduction of DCs interferes with tolerance, resulting in a stronger inflammatory response, and that other APC populations could compensate for the loss of immunogenic APC function in DC-depleted mice.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immune Tolerance/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation/immunology , Autoantigens/immunology , Autoimmunity/immunology , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , CD11c Antigen , Dendritic Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
8.
Curr Opin Urol ; 31(3): 228-235, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33742986

ABSTRACT

PURPOSE OF REVIEW: The aim of this review is to outline characteristics of the renal cell carcinoma (RCC) tumor immune microenvironment (TIME), the potential impact of tumor intrinsic alterations on the TIME and the value of metastatic tissue assessment in this context. RECENT FINDINGS: According to the latest European Association of Urology, European Society for Medical Oncology and National Comprehensive Cancer Network guidelines immune checkpoint inhibition represents a new core treatment strategy in advanced clear cell RCC (ccRCC). Despite its success, the prognosis of many RCC patients remains unsatisfactory most likely because of resistance mechanisms within the TIME. Moreover, most studies assess the primary tumor even though the advanced metastatic disease is targeted. Overall, metastatic RCC has hardly been investigated. First insights into the complexity of the genomic and immune landscape in RCC were recently provided. The functional impact of tumor intrinsic alterations on the TIME has just been described potentially contributing to therapy response in RCC. SUMMARY: The complexity of the RCC TIME and its potential interdependence with tumor intrinsic alterations has only just been recognized. A deeper understanding of the TIME may reveal predictive and prognostic biomarkers long-awaited in RCC, improve RCC patient stratification and could possibly be most instructive if assessed in metastatic tissue.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Biomarkers, Tumor , Humans , Kidney Neoplasms/therapy , Prognosis , Tumor Microenvironment
9.
Nature ; 494(7437): 361-5, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23376950

ABSTRACT

Cancer control by adaptive immunity involves a number of defined death and clearance mechanisms. However, efficient inhibition of exponential cancer growth by T cells and interferon-γ (IFN-γ) requires additional undefined mechanisms that arrest cancer cell proliferation. Here we show that the combined action of the T-helper-1-cell cytokines IFN-γ and tumour necrosis factor (TNF) directly induces permanent growth arrest in cancers. To safely separate senescence induced by tumour immunity from oncogene-induced senescence, we used a mouse model in which the Simian virus 40 large T antigen (Tag) expressed under the control of the rat insulin promoter creates tumours by attenuating p53- and Rb-mediated cell cycle control. When combined, IFN-γ and TNF drive Tag-expressing cancers into senescence by inducing permanent growth arrest in G1/G0, activation of p16INK4a (also known as CDKN2A), and downstream Rb hypophosphorylation at serine 795. This cytokine-induced senescence strictly requires STAT1 and TNFR1 (also known as TNFRSF1A) signalling in addition to p16INK4a. In vivo, Tag-specific T-helper 1 cells permanently arrest Tag-expressing cancers by inducing IFN-γ- and TNFR1-dependent senescence. Conversely, Tnfr1(-/-)Tag-expressing cancers resist cytokine-induced senescence and grow aggressively, even in TNFR1-expressing hosts. Finally, as IFN-γ and TNF induce senescence in numerous murine and human cancers, this may be a general mechanism for arresting cancer progression.


Subject(s)
Cellular Senescence/immunology , Cytokines/immunology , Neoplasms/immunology , Neoplasms/pathology , Th1 Cells/immunology , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Cell Cycle , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Disease Progression , Female , Humans , Interferon-gamma/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Oncogenes/genetics , Phosphoserine/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism , STAT1 Transcription Factor/metabolism , Time Factors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/immunology , Tumor Suppressor Protein p53/metabolism
10.
Am J Pathol ; 187(11): 2558-2569, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28822802

ABSTRACT

Activated leukocyte cell adhesion molecule (ALCAM) is expressed on various cell types, including leukocytes, endothelial cells, and certain tumor cells. Although ALCAM expression on tumor cells has been linked to tumor invasion and metastatic spread, the contribution of ALCAM expressed in cells forming the tumor stroma to cancer progression has not been investigated. In this study, ALCAM-deficient (ALCAM-/-) mice were used to evaluate the role of ALCAM in lung tumor growth and metastasis. ALCAM-/- mice displayed an altered blood vascular network in the lung and the diaphragm, indicative of an angiogenetic defect. The absence of ALCAM expression in cells forming the stromal tumor microenvironment profoundly affected lung tumor growth in three different i.v. metastasis models. In the case of Lewis lung carcinoma (LLC), an additional defect in tumor cell homing to the lungs and a resulting reduction in the number of lung tumor nodules were observed. Similarly, when LLC cells were implanted subcutaneously for the study of spontaneous tumor cell metastasis, the rate of LLC metastasis to the lungs was profoundly reduced in ALCAM-/- mice. Taken together, our work demonstrates for the first time the in vivo contribution of ALCAM to angiogenesis and reveals a novel role of stromally expressed ALCAM in supporting tumor growth and metastatic spread.


Subject(s)
Activated-Leukocyte Cell Adhesion Molecule/metabolism , Leukocytes/metabolism , Lung Neoplasms/pathology , Animals , Cell Adhesion/physiology , Cell Line, Tumor , Cell Proliferation , Lung Neoplasms/metabolism , Melanoma/metabolism , Mice, Inbred C57BL , Neoplasm Metastasis , Skin Neoplasms/pathology , Tumor Microenvironment/physiology
11.
Immunity ; 29(6): 934-46, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-19062317

ABSTRACT

Steady-state dendritic cells (DCs) maintain peripheral T cell tolerance, whereas mature DCs generate immunity. CD70 is a costimulatory ligand acquired upon DC maturation. To determine its impact on T cell fate, we have generated mice that constitutively express CD70 in conventional DCs (cDCs). In these mice, naive CD4+ and CD8+ T cells spontaneously convert into effector cells. Administration of peptide without adjuvant, which is ordinarily tolerogenic, elicited tumor-eradicating CD8+ T cell responses and robust CD4+ T cell-independent memory. CD70 was also constitutively expressed in cDCs that inducibly present viral epitopes. In this case, tolerance induction was prevented as well. The antigen-presenting DCs generated protective immunity to virus infection and broke a pre-existing state of CD8+ T cell tolerance. Thus, the sole expression of CD70 by otherwise immature cDCs sufficed to convert CD8+ T cell tolerance into immunity, defining the importance of CD27-CD70 interactions at the interface between T cell and DC.


Subject(s)
CD27 Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immune Tolerance/immunology , Neoplasms/immunology , Animals , CD27 Ligand/genetics , CD27 Ligand/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Dendritic Cells/metabolism , Immunologic Memory , Melanoma/immunology , Mice , Mice, Knockout , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
12.
J Immunol ; 189(2): 558-66, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22685313

ABSTRACT

Radiotherapy is an important treatment for cancer. The main mode of action is thought to be the irreversible damage to tumor cell DNA, but there is evidence that irradiation mobilizes tumor-specific immunity, and recent studies showed that the efficacy of high-dose radiotherapy depends on the presence of CD8(+) T cells. We show in this study that the efficacy of radiotherapy given as a single, high dose (10 Gy) crucially depends on dendritic cells and CD8(+) T cells, whereas CD4(+) T cells or macrophages are dispensable. We show that local high-dose irradiation results in activation of tumor-associated dendritic cells that in turn support tumor-specific effector CD8(+) T cells, thus identifying the mechanism that underlies radiotherapy-induced mobilization of tumor-specific immunity. We propose that in the absence of irradiation, the activation status of dendritic cells rather than the amount of tumor-derived Ag is the bottleneck, which precludes efficient anti-tumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/radiotherapy , Cell Differentiation/immunology , Dendritic Cells/immunology , Lung Neoplasms/immunology , Lung Neoplasms/radiotherapy , Animals , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/radiation effects , Carcinoma, Lewis Lung/pathology , Cell Differentiation/radiation effects , Dendritic Cells/metabolism , Dendritic Cells/radiation effects , Dose-Response Relationship, Radiation , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic
13.
Commun Med (Lond) ; 4(1): 5, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38182879

ABSTRACT

BACKGROUND: Tertiary lymphoid structures (TLSs) are dense accumulations of lymphocytes in inflamed peripheral tissues, including cancer, and are associated with improved survival and response to immunotherapy in various solid tumors. Histological TLS quantification has been proposed as a novel predictive and prognostic biomarker, but lack of standardized methods of TLS characterization hampers assessment of TLS densities across different patients, diseases, and clinical centers. METHODS: We introduce an approach based on HookNet-TLS, a multi-resolution deep learning model, for automated and unbiased TLS quantification and identification of germinal centers in routine hematoxylin and eosin stained digital pathology slides. We developed HookNet-TLS using n = 1019 manually annotated TCGA slides from clear cell renal cell carcinoma, muscle-invasive bladder cancer, and lung squamous cell carcinoma. RESULTS: Here we show that HookNet-TLS automates TLS quantification across multiple cancer types achieving human-level performance and demonstrates prognostic associations similar to visual assessment. CONCLUSIONS: HookNet-TLS has the potential to be used as a tool for objective quantification of TLS in routine H&E digital pathology slides. We make HookNet-TLS publicly available to promote its use in research.


Tertiary lymphoid structures (TLS) are dense accumulations of immune cells within a cancer. They have been associated with patient survival and treatment effectiveness. Quantification of TLS in cancer microscopy images may therefore aid clinical decision-making. However, no consensus for defining TLS in such images exists leading to inconsistent and variable findings across different labs and studies. We developed a computational tool for automated and objective TLS quantification in cancer images. The tool, called HookNet-TLS, integrates information from multiple image resolutions, which resembles the process of how a pathologist would identify these structures using a microscope. HookNet-TLS detected TLS similarly to trained researchers in three different tumor types. We provided access to HookNet-TLS to facilitate its development and use for TLS assessment in clinical decision-making and research into the role of TLS in cancer.

14.
J Virol ; 86(18): 9782-93, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22761378

ABSTRACT

The induction of strong CD8(+) T-cell responses against infectious diseases and cancer has remained a major challenge. Depending on the source of antigen and the infectious agent, priming of CD8(+) T cells requires direct and/or cross-presentation of antigenic peptides on major histocompatibility complex (MHC) class I molecules by professional antigen-presenting cells (APCs). However, both pathways show distinct preferences concerning antigen stability. Whereas direct presentation was shown to efficiently present peptides derived from rapidly degraded proteins, cross-presentation is dependent on long-lived antigen species. In this report, we analyzed the role of antigen stability on DNA vaccination and recombinant vaccinia virus (VV) infection using altered versions of the same antigen. The long-lived nucleoprotein (NP) of lymphocytic choriomeningitis virus (LCMV) can be targeted for degradation by N-terminal fusion to ubiquitin or, as we show here, to the ubiquitin-like modifier FAT10. Direct presentation by cells either transfected with NP-encoding plasmids or infected with recombinant VV in vitro was enhanced in the presence of short-lived antigens. In vivo, however, the highest induction of NP-specific CD8(+) T-cell responses was achieved in the presence of long-lived NP. Our experiments provide evidence that targeting antigens for proteasomal degradation does not improve the immunogenicity of DNA vaccines and recombinant VVs. Rather, it is the long-lived antigen that is superior for the efficient activation of MHC class I-restricted immune responses in vivo. Hence, our results suggest a dominant role for antigen cross-priming in DNA vaccination and recombinant VV infection.


Subject(s)
Antigens, Viral/metabolism , CD8-Positive T-Lymphocytes/immunology , Vaccines, DNA/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , Animals , Antigen Presentation , Antigens, Viral/genetics , Base Sequence , Cell Line , Cross Reactions , DNA, Viral/genetics , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/metabolism , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nucleoproteins/genetics , Nucleoproteins/immunology , Nucleoproteins/metabolism , Protein Stability , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Ubiquitin/genetics , Ubiquitin/immunology , Ubiquitin/metabolism , Ubiquitins/genetics , Ubiquitins/immunology , Ubiquitins/metabolism , Vaccines, DNA/genetics , Vaccinia virus/pathogenicity , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism
15.
Cancer Immun ; 13: 12, 2013.
Article in English | MEDLINE | ID: mdl-23882157

ABSTRACT

During cancer progression, malignant cells may evade immunosurveillance. However, evidence for immunological escape in humans is scarce. We report here the clinical course of a melanoma patient whose initial tumor was positive for the antigens NY-ESO-1, MAGE-C1, and Melan-A. Upon immunization with a recombinant vaccinia/fowlpox NY-ESO-1 construct, the patient experienced a mixed clinical response and spreading of the NY-ESO-1 epitopes in the CD4+ T cell compartment. After NY-ESO-1 protein + CpG immunization, the patient's anti-NY-ESO-1 IgG response increased. Over the following years, progressing lesions were resected and found to be NY-ESO-1-negative while being positive for MAGE-C1, Melan-A, and MHC-I. The fatal, inoperable brain metastasis was analyzed after his death and also proved to be NY-ESO-1-negative, while being positive for MAGE-C1 and Melan-A, as well as MHC-I. We propose that cancer control and cancer escape in this patient were governed by NY-ESO-1-specific immunological pressure. Our findings provide evidence for the existence of immunoediting and immunoescape in this cancer patient.


Subject(s)
Antigens, Neoplasm/immunology , Melanoma/immunology , Membrane Proteins/immunology , Humans , Immunohistochemistry , Male , Melanoma/pathology , Melanoma/secondary , Middle Aged
16.
Cancer Immun ; 13: 3, 2013.
Article in English | MEDLINE | ID: mdl-23390374

ABSTRACT

We investigated whether antibodies against intracellular tumor-associated antigens support tumor-specific immunity when administered together with a treatment that destroys the tumor. We propose that released antigens form immune complexes with the antibodies, which are then efficiently taken up by dendritic cells. We cloned the first human monoclonal antibodies against the Cancer/Testis (CT) antigen, NY-ESO-1. We tested whether the monoclonal anti-NY-ESO-1 antibody (12D7) facilitates cross-presentation of a NY-ESO-1-derived epitope by dendritic cells to human CD8+ T cells, and whether this results in the maturation of dendritic cells in vitro. We investigated the efficacy of 12D7 in combination with chemotherapy using BALB/c mice bearing syngeneic CT26 tumors that express intracellular NY-ESO-1. Human dendritic cells that were incubated with NY-ESO-1:12D7 immune complexes efficiently stimulated NY-ESO-1(157-165)/HLA-A2-specific human CD8+ T cells to produce interferon-γ, whereas NY-ESO-1 alone did not. Furthermore, the incubation of dendritic cells with NY-ESO-1:12D7 immune complexes resulted in the maturation of dendritic cells. Treatment of BALB/c mice that bear CT26/NY-ESO-1 tumors with 5-fluorouracil (5-FU) plus 12D7 was significantly more effective than chemotherapy alone. We propose systemic injection of monoclonal antibodies (mAbs) against tumor-associated antigens plus a treatment that promotes the local release of those antigens resulting in immune complex formation as a novel therapeutic modality for cancer.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Membrane Proteins/immunology , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Cell Differentiation/drug effects , Cloning, Molecular , Cross-Priming/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Epitope Mapping , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Humans , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/pathology , Treatment Outcome
17.
J Transl Med ; 11: 187, 2013 Aug 12.
Article in English | MEDLINE | ID: mdl-23937772

ABSTRACT

INTRODUCTION: Malignant pleural mesothelioma (MPM) is an incurable malignant disease, which results from chronic exposition to asbestos in at least 70% of the cases. Fibroblast activation protein (FAP) is predominantly expressed on the surface of reactive tumor-associated fibroblasts as well as on particular cancer types. Because of its expression on the cell surface, FAP is an attractive target for adoptive T cell therapy. T cells can be re-directed by retroviral transfer of chimeric antigen receptors (CAR) against tumor-associated antigens (TAA) and therefore represent a therapeutic strategy of adoptive immunotherapy. METHODS: To evaluate FAP expression immunohistochemistry was performed in tumor tissue from MPM patients. CD8+ human T cells were retrovirally transduced with an anti-FAP-F19-∆CD28/CD3ζ-CAR. T cell function was evaluated in vitro by cytokine release and cytotoxicity assays. In vivo function was tested with an intraperitoneal xenograft tumor model in immunodeficient mice. RESULTS: FAP was found to be expressed in all subtypes of MPM. Additionally, FAP expression was evaluated in healthy adult tissue samples and was only detected in specific areas in the pancreas, the placenta and very weakly for cervix and uterus. Expression of the anti-FAP-F19-∆CD28/CD3ζ-CAR in CD8+ T cells resulted in antigen-specific IFNγ release. Additionally, FAP-specific re-directed T cells lysed FAP positive mesothelioma cells and inflammatory fibroblasts in an antigen-specific manner in vitro. Furthermore, FAP-specific re-directed T cells inhibited the growth of FAP positive human tumor cells in the peritoneal cavity of mice and significantly prolonged survival of mice. CONCLUSION: FAP re-directed CD8+ T cells showed antigen-specific functionality in vitro and in vivo. Furthermore, FAP expression was verified in all MPM histotypes. Therefore, our data support performing a phase I clinical trial in which MPM patients are treated with adoptively transferred FAP-specific re-directed T cells.


Subject(s)
Gelatinases/metabolism , Immunotherapy , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Membrane Proteins/metabolism , Mesothelioma/immunology , Mesothelioma/therapy , Pleural Neoplasms/immunology , Pleural Neoplasms/therapy , Serine Endopeptidases/metabolism , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cytotoxicity, Immunologic , Endopeptidases , Humans , Immunohistochemistry , Mesothelioma, Malignant , Mice , Peritoneum/pathology , Recombinant Proteins/metabolism , Stromal Cells/metabolism , T-Lymphocytes/metabolism , Transduction, Genetic , Xenograft Model Antitumor Assays
18.
Proc Natl Acad Sci U S A ; 107(1): 199-203, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-20018763

ABSTRACT

Peripheral T-cell tolerance is thought to significantly contribute to the prevention of autoimmunity, and it has been shown that antigen-presenting steady-state dendritic cells efficiently induce peripheral tolerance. We previously showed that dendritic-cell-induced tolerance is a T-cell-intrinsic process that depends on coinhibitory molecules such as programmed death-1. Here we specifically analyze the involvement of FoxP3(+) regulatory T cells, which are known to be important for maintenance of self-tolerance. We show that antigen presentation by steady-state dendritic cells failed to induce peripheral tolerance in the absence of FoxP3(+) regulatory T cells but induced protective CD8(+) T-cell-mediated immunity instead. Regulatory T-cell-depleted mice had massively increased numbers of dendritic cells in lymph nodes. Dendritic cells isolated from mice without regulatory T cells had up-regulated costimulatory molecules and showed stronger T-cell stimulatory capacity ex vivo, suggesting that regulatory T cells contribute to peripheral tolerance by keeping the dendritic cells in an immature state. Using blocking antibodies, we demonstrate that CTLA-4 but not IL-10 is necessary for control of dendritic cells by regulatory T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Forkhead Transcription Factors/immunology , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD/immunology , CTLA-4 Antigen , Forkhead Transcription Factors/genetics , Interleukin-10/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
19.
Proc Natl Acad Sci U S A ; 107(34): 15187-92, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20696919

ABSTRACT

Cancer/testis (CT) antigens represent prime candidates for immunotherapy in cancer patients, because their expression is restricted to cancer cells and germ cells of the testis. MAGE-C1/CT7 is a CT antigen that is highly expressed in several types of cancers. Spontaneous occurrence of CT7-specific antibodies was previously detected by SEREX screen in a melanoma patient. However, naturally occurring CT7-specific T-cell responses have thus far not been detected. Peripheral blood mononuclear cells (PBMCs) from 26 metastatic melanoma patients expressing CT7 in their tumor lesions (CT7(+)) were analyzed for CT7-specific T-cell responses using overlapping peptides. CT7-specific CD4(+) T-cell responses were detected in three patients (11.5%). These CT7-specific CD4(+) T-cell responses were detectable in melanoma patients' PBMCs exclusively from preexisting CD45RA(-) memory CD4(+) T-cell pool. Additional CT7-specific memory CD4(+) T-cell responses were detected in CT7(+) melanoma patients after depletion of CD4(+)CD25high Treg cells showing that Treg cells impact on CT7-specific CD4(+) T cells in melanoma patients. CT7-specific CD4(+) T-cell clones were generated and used to define minimal epitopes, restriction elements, and confirm the recognition of naturally processed antigen. Surprisingly, these clones were able to secrete perforin and exert cytotoxicity. This study shows that CT7 can induce specific cellular immunity in melanoma patients. Based on these findings, CT7 will be further explored as a potential vaccine for melanoma immunotherapy.


Subject(s)
Antigens, Neoplasm/metabolism , Melanoma/immunology , Neoplasm Proteins/metabolism , Amino Acid Sequence , Antigens, Neoplasm/genetics , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Epitope Mapping , Epitopes/genetics , Humans , Immunologic Memory , In Vitro Techniques , Melanoma/genetics , Melanoma/secondary , Melanoma/therapy , Neoplasm Proteins/genetics , T-Lymphocytes, Regulatory/immunology
20.
Nat Rev Nephrol ; 19(8): 525-537, 2023 08.
Article in English | MEDLINE | ID: mdl-37046081

ABSTRACT

Tertiary lymphoid structures (TLSs) are ectopic lymphoid tissues that drive antigen-specific immune responses at sites of chronic inflammation. Unlike secondary lymphoid organs such as lymph nodes, TLSs lack capsules and have their own unique characteristics and functions. The presumed influence of TLSs on the disease course has led to widespread interest in obtaining a better understanding of their biology and function. Studies using single-cell analyses have suggested heterogeneity in TLS composition and phenotype, and consequently, functional correlates with disease progression are sometimes conflicting. The presence of TLSs correlates with a favourable disease course in cancer and infection. Conversely, in autoimmune diseases and chronic age-related inflammatory diseases including chronic kidney disease, the presence of TLSs is associated with a more severe disease course. However, the detailed mechanisms that underlie these clinical associations are not fully understood. To what extent the mechanisms of TLS development and maturation are shared across organs and diseases is also still obscure. Improved understanding of TLS development and function at the cellular and molecular levels may enable the exploitation of these structures to improve therapies for chronic diseases, including chronic kidney disease.


Subject(s)
Neoplasms , Renal Insufficiency, Chronic , Tertiary Lymphoid Structures , Humans , Tertiary Lymphoid Structures/genetics , Tertiary Lymphoid Structures/pathology , Disease Progression , Chronic Disease
SELECTION OF CITATIONS
SEARCH DETAIL