Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
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.

2.
Nat Commun ; 14(1): 5154, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37620318

ABSTRACT

Immune checkpoint inhibitor treatment has the potential to prolong survival in non-small cell lung cancer (NSCLC), however, some of the patients develop resistance following initial response. Here, we analyze the immune phenotype of matching tumor samples from a cohort of NSCLC patients showing good initial response to immune checkpoint inhibitors, followed by acquired resistance at later time points. By using imaging mass cytometry and whole exome and RNA sequencing, we detect two patterns of resistance¨: One group of patients is characterized by reduced numbers of tumor-infiltrating CD8+ T cells and reduced expression of PD-L1 after development of resistance, whereas the other group shows high CD8+ T cell infiltration and high expression of PD-L1 in addition to markedly elevated expression of other immune-inhibitory molecules. In two cases, we detect downregulation of type I and II IFN pathways following progression to resistance, which could lead to an impaired anti-tumor immune response. This study thus captures the development of immune checkpoint inhibitor resistance as it progresses and deepens our mechanistic understanding of immunotherapy response in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , CD8-Positive T-Lymphocytes , B7-H1 Antigen/genetics , Immune Checkpoint Inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Immunosuppressive Agents , Phenotype
3.
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
4.
J Immunother Cancer ; 10(6)2022 06.
Article in English | MEDLINE | ID: mdl-35688559

ABSTRACT

BACKGROUND: Dendritic cells (DCs) are professional antigen presenting cells that initiate immune defense to pathogens and tumor cells. Human tumors contain only few DCs that mostly display a non-activated phenotype. Hence, activation of tumor-associated DCs may improve efficacy of cancer immunotherapies. Toll-like receptor (TLR) agonists and interferons are known to promote DC maturation. However, it is unclear if DCs in human tumors respond to activation signals and which stimuli induce the optimal activation of human tumor DCs. METHODS: We first screened combinations of TLR agonists, a STING agonist and interferons (IFNs) for their ability to activate human conventional DCs (cDCs). Two combinations: TL8-506 (a TLR8 agonist)+IFN-γ and TL8-506+Poly(I:C) (a TLR3 agonist) were studied in more detail. cDC1s and cDC2s derived from cord blood stem cells, blood or patient tumor samples were stimulated with either TL8-506+IFN-γ or TL8-506+Poly(I:C). Different activation markers were analyzed by ELISA, flow cytometry, NanoString nCounter Technology or single-cell RNA-sequencing. T cell activation and migration assays were performed to assess functional consequences of cDC activation. RESULTS: We show that TL8-506 synergized with IFN-γ or Poly(I:C) to induce high expression of different chemokines and cytokines including interleukin (IL)-12p70 in human cord blood and blood cDC subsets in a combination-specific manner. Importantly, both combinations induced the activation of cDC subsets in patient tumor samples ex vivo. The expression of immunostimulatory genes important for anticancer responses including CD40, IFNB1, IFNL1, IL12A and IL12B were upregulated on stimulation. Furthermore, chemokines associated with CD8+ T cell recruitment were induced in tumor-derived cDCs in response to TL8-506 combinations. In vitro activation and migration assays confirmed that stimulated cDCs induce T cell activation and migration. CONCLUSIONS: Our data suggest that cord blood-derived and blood-derived cDCs are a good surrogate to study treatment responses in human tumor cDCs. While most cDCs in human tumors display a non-activated phenotype, TL8-506 combinations drive human tumor cDCs towards an immunostimulatory phenotype associated with Th1 responses on stimulation. Hence, TL8-506-based combinations may be promising candidates to initiate or boost antitumor responses in patients with cancer.


Subject(s)
Neoplasms , Toll-Like Receptor 8 , Adjuvants, Immunologic/pharmacology , Cytokines/metabolism , Dendritic Cells , Humans , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Interleukin-12/metabolism , Poly I-C/metabolism , Poly I-C/pharmacology
5.
Trends Cancer ; 8(2): 145-157, 2022 02.
Article in English | MEDLINE | ID: mdl-34815204

ABSTRACT

Metastasis is an intricate process whereby tumor cells migrate from the primary tumor, survive in the circulation, seed distal organs, and proliferate to create metastatic foci. CD8+ T cells can detect and eliminate tumor cells. Research on CD8+ T cell-dependent antitumor immunity has classically focused on its role in the primary tumor. There is increasing evidence, however, that CD8+ T cells have unique antimetastatic functions in various steps of the metastatic cascade. Here, we review the mechanisms whereby CD8+ T cells control metastatic lesions. We discuss their role in each step of metastasis, metastatic dormancy, and metastatic clonal evolution as well as the consequent clinical repercussions.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , Neoplasms/pathology
6.
Sci Transl Med ; 13(611): eabc8188, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34524861

ABSTRACT

Platelet-derived peripheral serotonin has pleiotropic effects on coagulation, metabolism, tissue regeneration, and cancer growth; however, the effect of serotonin on the tumor microenvironment remains understudied. Peripheral serotonin­deficient (Tph1−/−) mice displayed reduced growth of subcutaneous and orthotopically injected syngeneic murine pancreatic and colorectal cancers with enhanced accumulation of functional CD8+ T cells compared to control C57BL/6 mice, resulting in extended overall survival. Subcutaneous and orthotopic syngeneic tumors from Tph1−/− mice expressed less programmed cell death 1 ligand 1 (PD-L1), suggesting serotonin-mediated regulation. Serotonin enhanced expression of PD-L1 on mouse and human cancer cells in vitro via serotonylation, which is the formation of covalent bonds between glutamine residues and serotonin, resulting in activation of small G proteins. Serotonin concentrations in metastases of patients with abdominal tumors negatively correlated to the number of CD8+ tumor-infiltrating T cells. Depletion of serotonin cargo or inhibition of serotonin release from thrombocytes decreased growth of syngeneic pancreatic and colorectal tumors in wild-type mice, increased CD8+ T cell influx, and decreased PD-L1 expression. Pharmacological serotonin depletion with oral fluoxetine or intraperitoneal injection of the TPH1 inhibitor telotristat augmented the effects of programmed cell death protein 1 (PD-1) checkpoint blockade and triggered long-term tumor control in mice subcutaneously inoculated with syngeneic colorectal and pancreatic tumors. Overall, peripheral serotonin weakens effector functions of CD8+ T cells within tumors. Clinically approved serotonin targeting agents alone or in combination with PD-1 blockade provided long-term control of established tumors in murine models, warranting further investigation of the clinical translatability of these findings.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Animals , Mice , Neoplasms/drug therapy , Serotonin
7.
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
8.
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
9.
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
10.
Nat Commun ; 12(1): 769, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33536445

ABSTRACT

Some breast tumors metastasize aggressively whereas others remain dormant for years. The mechanism governing metastatic dormancy remains largely unknown. Through high-parametric single-cell mapping in mice, we identify a discrete population of CD39+PD-1+CD8+ T cells in primary tumors and in dormant metastasis, which is hardly found in aggressively metastasizing tumors. Using blocking antibodies, we find that dormancy depends on TNFα and IFNγ. Immunotherapy reduces the number of dormant cancer cells in the lungs. Adoptive transfer of purified CD39+PD-1+CD8+ T cells prevents metastatic outgrowth. In human breast cancer, the frequency of CD39+PD-1+CD8+ but not total CD8+ T cells correlates with delayed metastatic relapse after resection (disease-free survival), thus underlining the biological relevance of CD39+PD-1+CD8+ T cells for controlling experimental and human breast cancer. Thus, we suggest that a primary breast tumor could prime a systemic, CD39+PD-1+CD8+ T cell response that favors metastatic dormancy in the lungs.


Subject(s)
Antigens, CD/immunology , Apyrase/immunology , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Mammary Neoplasms, Experimental/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Antigens, CD/metabolism , Apyrase/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Humans , Immunotherapy , Lung/immunology , Lung/pathology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Knockout , Neoplasm Metastasis , Programmed Cell Death 1 Receptor/metabolism
11.
Front Immunol ; 12: 793964, 2021.
Article in English | MEDLINE | ID: mdl-34987518

ABSTRACT

Candidate immune biomarkers have been proposed for predicting response to immunotherapy in urothelial cancer (UC). Yet, these biomarkers are imperfect and lack predictive power. A comprehensive overview of the tumor immune contexture, including Tertiary Lymphoid structures (TLS), is needed to better understand the immunotherapy response in UC. We analyzed tumor sections by quantitative multiplex immunofluorescence to characterize immune cell subsets in various tumor compartments in tumors without pretreatment and tumors exposed to preoperative anti-PD1/CTLA-4 checkpoint inhibitors (NABUCCO trial). Pronounced immune cell presence was found in UC invasive margins compared to tumor and stroma regions. CD8+PD1+ T-cells were present in UC, particularly following immunotherapy. The cellular composition of TLS was assessed by multiplex immunofluorescence (CD3, CD8, FoxP3, CD68, CD20, PanCK, DAPI) to explore specific TLS clusters based on varying immune subset densities. Using a k-means clustering algorithm, we found five distinct cellular composition clusters. Tumors unresponsive to anti-PD-1/CTLA-4 immunotherapy showed enrichment of a FoxP3+ T-cell-low TLS cluster after treatment. Additionally, cluster 5 (macrophage low) TLS were significantly higher after pre-operative immunotherapy, compared to untreated tumors. We also compared the immune cell composition and maturation stages between superficial (submucosal) and deeper TLS, revealing that superficial TLS had more pronounced T-helper cells and enrichment of early TLS than TLS located in deeper tissue. Furthermore, superficial TLS displayed a lower fraction of secondary follicle like TLS than deeper TLS. Taken together, our results provide a detailed quantitative overview of the tumor immune landscape in UC, which can provide a basis for further studies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immune Checkpoint Inhibitors/therapeutic use , Ipilimumab/therapeutic use , Nivolumab/therapeutic use , T-Lymphocytes, Helper-Inducer/immunology , Urothelium/metabolism , Aged , CTLA-4 Antigen/antagonists & inhibitors , Cell Differentiation , Cells, Cultured , Female , Fluorescent Antibody Technique , Forkhead Transcription Factors/metabolism , Humans , Immunotherapy , Lymphocyte Activation , Male , Middle Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Tertiary Lymphoid Structures , Treatment Outcome , Tumor Microenvironment , Urologic Neoplasms , Urothelium/pathology
12.
Front Immunol ; 12: 793992, 2021.
Article in English | MEDLINE | ID: mdl-35145509

ABSTRACT

Lymphoid neogenesis gives rise to tertiary lymphoid structures (TLS) in the periphery of multiple cancer types including muscle invasive bladder cancer (MIBC) where it has positive prognostic and predictive associations. Here, we explored molecular, clinical, and histological data of The Cancer Genome Atlas, as well as the IMvigor210 dataset to study factors associated with TLS development and function in the tumor microenvironment (TME) of MIBC. We also analyzed tumor immune composition including TLS in an independent, retrospective MIBC cohort. We found that the combination of TLS density and tumor mutational burden provides a novel independent prognostic biomarker in MIBC. Gene expression profiles obtained from intratumoral regions that rarely contain TLS in MIBC showed poor correlation with the prognostic TLS density measured in tumor periphery. Tumors with high TLS density showed increased gene signatures as well as infiltration of activated lymphocytes. Intratumoral B-cell and CD8+ T-cell co-infiltration was frequent in TLS-high samples, and such regions harbored the highest proportion of PD-1+TCF1+ progenitor-like T cells, naïve T cells, and activated B cells when compared to regions predominantly infiltrated by either B cells or CD8+ T cells alone. We found four TLS maturation subtypes; however, differences in TLS composition appeared to be dictated by the TME and not by the TLS maturation status. Finally, we identified one downregulated and three upregulated non-immune cell-related genes in TME with high TLS density, which may represent candidates for tumor-intrinsic regulation of lymphoid neogenesis. Our study provides novel insights into TLS-associated gene expression and immune contexture of MIBC and indicates towards the relevance of B-cell and CD8+ T-cell interactions in anti-tumor immunity within and outside TLS.


Subject(s)
Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Muscles/pathology , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/metabolism , Urinary Bladder Neoplasms/pathology , Biomarkers , Biomarkers, Tumor , Diagnostic Imaging , Disease Susceptibility , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation , Humans , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating , Microvascular Density , Neoplasm Invasiveness , Neoplasm Staging , Neovascularization, Pathologic/diagnostic imaging , Prognosis , Tumor Microenvironment , Urinary Bladder Neoplasms/etiology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/mortality
13.
Nat Med ; 26(12): 1839-1844, 2020 12.
Article in English | MEDLINE | ID: mdl-33046870

ABSTRACT

Preoperative immunotherapy with anti-PD1 plus anti-CTLA4 antibodies has shown remarkable pathological responses in melanoma1 and colorectal cancer2. In NABUCCO (ClinicalTrials.gov: NCT03387761 ), a single-arm feasibility trial, 24 patients with stage III urothelial cancer (UC) received two doses of ipilimumab and two doses of nivolumab, followed by resection. The primary endpoint was feasibility to resect within 12 weeks from treatment start. All patients were evaluable for the study endpoints and underwent resection, 23 (96%) within 12 weeks. Grade 3-4 immune-related adverse events occurred in 55% of patients and in 41% of patients when excluding clinically insignificant laboratory abnormalities. Eleven patients (46%) had a pathological complete response (pCR), meeting the secondary efficacy endpoint. Fourteen patients (58%) had no remaining invasive disease (pCR or pTisN0/pTaN0). In contrast to studies with anti-PD1/PD-L1 monotherapy, complete response to ipilimumab plus nivolumab was independent of baseline CD8+ presence or T-effector signatures. Induction of tertiary lymphoid structures upon treatment was observed in responding patients. Our data indicate that combined CTLA-4 plus PD-1 blockade might provide an effective preoperative treatment strategy in locoregionally advanced UC, irrespective of pre-existing CD8+ T cell activity.


Subject(s)
Ipilimumab/administration & dosage , Neoplasms/drug therapy , Nivolumab/administration & dosage , Urothelium/pathology , Adult , Aged , Antibodies, Monoclonal , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Female , Humans , Male , Middle Aged , Neoplasm Staging , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/surgery , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Urothelium/drug effects , Urothelium/immunology , Urothelium/surgery
14.
Leukemia ; 34(10): 2688-2703, 2020 10.
Article in English | MEDLINE | ID: mdl-32358567

ABSTRACT

Acute myeloid leukemia (AML) initiating and sustaining cells maintain high cell-surface similarity with their cells-of-origin, i.e., hematopoietic stem and progenitor cells (HSPCs), and identification of truly distinguishing leukemia-private antigens has remained elusive to date. To nonetheless utilize surface antigen-directed immunotherapy in AML, we here propose targeting both, healthy and malignant human HSPC, by chimeric antigen receptor (CAR) T-cells with specificity against CD117, the cognate receptor for stem cell factor. This approach should spare most mature hematopoietic cells and would require CAR T termination followed by subsequent transplantation of healthy HSPCs to rescue hematopoiesis. We successfully generated anti-CD117 CAR T-cells from healthy donors and AML patients. Anti-CD117 CAR T-cells efficiently targeted healthy and leukemic CD117-positive cells in vitro. In mice xenografted with healthy human hematopoiesis, they eliminated CD117-expressing, but not CD117-negative human cells. Importantly, in mice xenografted with primary human CD117-positive AML, they eradicated disease in a therapeutic setting. Administration of ATG in combination with rituximab, which binds to the co-expressed CAR T-cell transduction/selection marker RQR8, led to CAR T-cell depletion. Thus, we here provide the first proof of concept for the generation and preclinical efficacy of CAR T-cells directed against CD117-expressing human hematopoietic cells.


Subject(s)
Immunotherapy, Adoptive , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Biomarkers , Biopsy , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Line, Tumor , Disease Models, Animal , Gene Expression , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/immunology , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/therapy , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Lymphocyte Depletion , Mice , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Treatment Outcome , Xenograft Model Antitumor Assays
15.
Cell Rep ; 29(5): 1236-1248.e7, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31665636

ABSTRACT

Sensing of cytoplasmic DNA by cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) results in production of the dinucleotide cGAMP and consecutive activation of stimulator of interferon genes (STING) followed by production of type I interferon (IFN). Although cancer cells contain supra-normal concentrations of cytoplasmic DNA, they rarely produce type I IFN spontaneously. This suggests that defects in the DNA-sensing pathway may serve as an immune escape mechanism. We find that cancer cells produce cGAMP that is transferred via gap junctions to tumor-associated dendritic cells (DCs) and macrophages, which respond by producing type I IFN in situ. Cancer-cell-intrinsic expression of cGAS, but not STING, promotes infiltration by effector CD8+ T cells and consequently results in prolonged survival. Furthermore, cGAS-expressing cancers respond better to genotoxic treatments and immunotherapy. Thus, cancer-cell-derived cGAMP is crucial to protective anti-tumor CD8+ T cell immunity. Consequently, cancer-cell-intrinsic expression of cGAS determines tumor immunogenicity and makes tumors hot. These findings are relevant for genotoxic and immune therapies for cancer.


Subject(s)
Neoplasms/immunology , Nucleotidyltransferases/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , DNA Damage , Dendritic Cells/metabolism , Disease Progression , Humans , Immunotherapy , Interferon Type I/metabolism , Membrane Proteins , Mice, Inbred C57BL , Microsatellite Repeats/genetics , Neoplasms/drug therapy , Neoplasms/pathology , Nucleotides, Cyclic/metabolism
16.
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
17.
Sci Transl Med ; 10(469)2018 11 28.
Article in English | MEDLINE | ID: mdl-30487251

ABSTRACT

Allogeneic hematopoietic cell transplantation (allo-HCT) not only is an effective treatment for several hematologic malignancies but can also result in potentially life-threatening graft-versus-host disease (GvHD). GvHD is caused by T cells within the allograft attacking nonmalignant host tissues; however, these same T cells mediate the therapeutic graft-versus-leukemia (GvL) response. Thus, there is an urgent need to understand how to mechanistically uncouple GvL from GvHD. Using preclinical models of full and partial MHC-mismatched HCT, we here show that the granulocyte-macrophage colony-stimulating factor (GM-CSF) produced by allogeneic T cells distinguishes between the two processes. GM-CSF drives GvHD pathology by licensing donor-derived phagocytes to produce inflammatory mediators such as interleukin-1ß and reactive oxygen species. In contrast, GM-CSF did not affect allogeneic T cells or their capacity to eliminate leukemic cells, retaining undiminished GvL responses. Last, tissue biopsies and peripheral blood mononuclear cells from patients with grade IV GvHD showed an elevation of GM-CSF-producing T cells, suggesting that GM-CSF neutralization has translational potential in allo-HCT.


Subject(s)
Graft vs Host Disease/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunity/drug effects , Leukemia/immunology , Myeloid Cells/metabolism , Animals , Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation , Histocompatibility Antigens/metabolism , Humans , Interferon-gamma/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transplantation, Homologous
18.
Methods Mol Biol ; 1845: 71-86, 2018.
Article in English | MEDLINE | ID: mdl-30141008

ABSTRACT

Tertiary lymphoid structures (TLS) develop in the human tumor microenvironment and correlate with prolonged survival in most cancer types. We recently demonstrated that TLS development follows sequential maturation stages and culminates in the generation of a germinal center (GC) reaction. This maturation process is crucial for the prognostic relevance of TLS in lung and colorectal cancer patients.The mechanisms underlying TLS development in various inflammatory conditions or their functional relevance in tumor immunity are not fully understood. Investigating which cell types and soluble mediators orchestrate lymphoid neogenesis in human tissues requires a method that allows simultaneous detection of multiple markers.Here, we describe a quantitative pathology approach to identify and quantify different TLS maturation stages in combination with other parameters. This approach consists of seven-color immunofluorescence protocol using tyramide signal amplification combined with multispectral microscopy and quantitative data acquisition from histological images.


Subject(s)
Neoplasms/pathology , Tertiary Lymphoid Structures/pathology , Tumor Microenvironment , Biomarkers , Fluorescent Antibody Technique , Germinal Center/immunology , Germinal Center/metabolism , Germinal Center/pathology , Humans , Image Processing, Computer-Assisted , Immunohistochemistry/methods , Microscopy, Fluorescence/methods , Neoplasm Staging , Neoplasms/immunology , Neoplasms/metabolism , Software , Tertiary Lymphoid Structures/immunology , Tertiary Lymphoid Structures/metabolism , Tumor Microenvironment/immunology
19.
Clin Cancer Res ; 24(24): 6345-6354, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30154226

ABSTRACT

PURPOSE: Combination of immune checkpoint inhibitors with chemotherapy is under investigation for cancer treatment. EXPERIMENTAL DESIGN: We studied the rationale of such a combination for treating mesothelioma, a disease with limited treatment options. RESULTS: The combination of gemcitabine and immune checkpoint inhibitors outperformed immunotherapy alone with regard to tumor control and survival in a preclinical mesothelioma model; however, the addition of dexamethasone to gemcitabine and immune checkpoint inhibitors nullified the synergistic clinical response. Furthermore, treatment with gemcitabine plus anti-PD-1 resulted in an objective clinical response in two patients with mesothelioma, who were resistant to gemcitabine or anti-PD-1 as monotherapy. CONCLUSIONS: Thus, treatment of mesothelioma with a combination of gemcitabine with immune checkpoint inhibitors is feasible and results in synergistic clinical response compared with single treatment in the absence of steroids.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Immunomodulation/drug effects , Lung Neoplasms/immunology , Mesothelioma/immunology , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Biopsy , Cell Line, Tumor , Deoxycytidine/pharmacology , Disease Models, Animal , Drug Synergism , Gene Expression , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mesothelioma/diagnosis , Mesothelioma/drug therapy , Mesothelioma/metabolism , Mesothelioma, Malignant , Mice , Positron Emission Tomography Computed Tomography , Prognosis , Treatment Outcome , Xenograft Model Antitumor Assays , Gemcitabine
20.
J Exp Med ; 215(8): 2055-2072, 2018 08 06.
Article in English | MEDLINE | ID: mdl-29970473

ABSTRACT

Eosinophils are predominantly known for their contribution to allergy. Here, we have examined the function and regulation of gastrointestinal eosinophils in the steady-state and during infection with Helicobacter pylori or Citrobacter rodentium We find that eosinophils are recruited to sites of infection, directly encounter live bacteria, and activate a signature transcriptional program; this applies also to human gastrointestinal eosinophils in humanized mice. The genetic or anti-IL-5-mediated depletion of eosinophils results in improved control of the infection, increased inflammation, and more pronounced Th1 responses. Eosinophils control Th1 responses via the IFN-γ-dependent up-regulation of PD-L1. Furthermore, we find that the conditional loss of IFN-γR in eosinophils phenocopies the effects of eosinophil depletion. Eosinophils further possess bactericidal properties that require their degranulation and the deployment of extracellular traps. Our results highlight two novel functions of this elusive cell type and link it to gastrointestinal homeostasis and anti-bacterial defense.


Subject(s)
Citrobacter rodentium/physiology , Eosinophils/immunology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Helicobacter pylori/physiology , Inflammation/immunology , Inflammation/microbiology , Th1 Cells/immunology , Acute Disease , Animals , Antibodies, Bacterial/immunology , Bacterial Infections/immunology , Bacterial Infections/microbiology , Bacterial Infections/pathology , Cell Degranulation , Cell Proliferation , Colitis/immunology , Colitis/microbiology , Colitis/pathology , Cytokines/biosynthesis , Disease Models, Animal , Eosinophils/physiology , Extracellular Traps/metabolism , Gastrointestinal Tract/immunology , Homeostasis , Immunity, Innate , Immunity, Mucosal , Inflammation/pathology , Interferon-gamma/metabolism , Mice, Inbred C57BL , Signal Transduction , Th17 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...