Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
J Transl Med ; 22(1): 292, 2024 Mar 19.
Article En | MEDLINE | ID: mdl-38504345

BACKGROUND: Naturally occurring colorectal cancers (CRC) in rhesus macaques share many features with their human counterparts and are useful models for cancer immunotherapy; but mechanistic data are lacking regarding the comparative molecular pathogenesis of these cancers. METHODS: We conducted state-of-the-art imaging including CT and PET, clinical assessments, and pathological review of 24 rhesus macaques with naturally occurring CRC. Additionally, we molecularly characterized these tumors utilizing immunohistochemistry (IHC), microsatellite instability assays, DNAseq, transcriptomics, and developed a DNA methylation-specific qPCR assay for MLH1, CACNA1G, CDKN2A, CRABP1, and NEUROG1, human markers for CpG island methylator phenotype (CIMP). We furthermore employed Monte-Carlo simulations to in-silico model alterations in DNA topology in transcription-factor binding site-rich promoter regions upon experimentally demonstrated DNA methylation. RESULTS: Similar cancer histology, progression patterns, and co-morbidities could be observed in rhesus as reported for human CRC patients. IHC identified loss of MLH1 and PMS2 in all cases, with functional microsatellite instability. DNA sequencing revealed the close genetic relatedness to human CRCs, including a similar mutational signature, chromosomal instability, and functionally-relevant mutations affecting KRAS (G12D), TP53 (R175H, R273*), APC, AMER1, ALK, and ARID1A. Interestingly, MLH1 mutations were rarely identified on a somatic or germline level. Transcriptomics not only corroborated the similarities of rhesus and human CRCs, but also demonstrated the significant downregulation of MLH1 but not MSH2, MSH6, or PMS2 in rhesus CRCs. Methylation-specific qPCR suggested CIMP-positivity in 9/16 rhesus CRCs, but all 16/16 exhibited significant MLH1 promoter hypermethylation. DNA hypermethylation was modelled to affect DNA topology, particularly propeller twist and roll profiles. Modelling the DNA topology of a transcription factor binding motif (TFAP2A) in the MLH1 promoter that overlapped with a methylation-specific probe, we observed significant differences in DNA topology upon experimentally shown DNA methylation. This suggests a role of transcription factor binding interference in epigenetic silencing of MLH1 in rhesus CRCs. CONCLUSIONS: These data indicate that epigenetic silencing suppresses MLH1 transcription, induces the loss of MLH1 protein, abrogates mismatch repair, and drives genomic instability in naturally occurring CRC in rhesus macaques. We consider this spontaneous, uninduced CRC in immunocompetent, treatment-naïve rhesus macaques to be a uniquely informative model for human CRC.


Brain Neoplasms , Colorectal Neoplasms , Microsatellite Instability , Neoplastic Syndromes, Hereditary , Humans , Animals , Macaca mulatta/genetics , Macaca mulatta/metabolism , MutL Protein Homolog 1/genetics , Mismatch Repair Endonuclease PMS2/genetics , Mismatch Repair Endonuclease PMS2/metabolism , Colorectal Neoplasms/pathology , DNA Methylation/genetics , Epigenesis, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism , DNA/metabolism , DNA Mismatch Repair/genetics
2.
Sci Transl Med ; 11(496)2019 06 12.
Article En | MEDLINE | ID: mdl-31189721

Endogenous costimulatory molecules on T cells such as 4-1BB (CD137) can be leveraged for cancer immunotherapy. Systemic administration of agonistic anti-4-1BB antibodies, although effective preclinically, has not advanced to phase 3 trials because they have been hampered by both dependency on Fcγ receptor-mediated hyperclustering and hepatotoxicity. To overcome these issues, we engineered proteins simultaneously targeting 4-1BB and a tumor stroma or tumor antigen: FAP-4-1BBL (RG7826) and CD19-4-1BBL. In the presence of a T cell receptor signal, they provide potent T cell costimulation strictly dependent on tumor antigen-mediated hyperclustering without systemic activation by FcγR binding. We could show targeting of FAP-4-1BBL to FAP-expressing tumor stroma and lymph nodes in a colorectal cancer-bearing rhesus monkey. Combination of FAP-4-1BBL with tumor antigen-targeted T cell bispecific (TCB) molecules in human tumor samples led to increased IFN-γ and granzyme B secretion. Further, combination of FAP- or CD19-4-1BBL with CEA-TCB (RG7802) or CD20-TCB (RG6026), respectively, resulted in tumor remission in mouse models, accompanied by intratumoral accumulation of activated effector CD8+ T cells. FAP- and CD19-4-1BBL thus represent an off-the-shelf combination immunotherapy without requiring genetic modification of effector cells for the treatment of solid and hematological malignancies.


Antibodies, Bispecific/metabolism , CD8-Positive T-Lymphocytes/metabolism , Antibodies, Bispecific/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cell Proliferation/physiology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Humans , Immunotherapy , Lymph Nodes/immunology , Lymph Nodes/metabolism , Neoplasms/immunology , Neoplasms/therapy
3.
J Immunother Cancer ; 7(1): 55, 2019 02 26.
Article En | MEDLINE | ID: mdl-30808414

BACKGROUND: Immunogenic radiotherapy (RT) can act synergistically with immune checkpoint blockers (ICBs). However, alternatives are needed for non-responding patients and those with pre-existing or ICB-induced autoimmune symptoms. Combination of RT with IL-2 could be an alternative. But IL-2 has a short half-life, and, by binding to its high-affinity receptor, it strongly stimulates immunosuppressive CD4+ Tregs and seems to promote potentially life-threatening vascular leakage. IL-2/anti-IL-2 complexes (IL-2c), which bind to the low-affinity receptor, have been reported to circumvent these disadvantages but they have not yet been thoroughly tested in conjunction with radiotherapy. METHODS: We evaluated, in three mouse models, the antitumoral effects induced by hypofractionated RT (hRT) plus IL-2c. We also used non-invasive imaging with a newly developed PET tracer based on therapeutically active IL-2c and a PD-L1 PET tracer for the theranostic evaluation of the treatment and its side effects. RESULTS: Treatment of mice bearing established B16 melanomas with hRT + IL-2c was superior to hRT + uncomplexed IL-2 or hRT alone; IL-2c alone was not effective. hRT + IL-2c was also synergistic in mice bearing C51 colon carcinomas or 4T1 mammary carcinomas. The better antitumor response correlated with increased tumor-specific CD8+ T cells and NK cells, but not CD4+ Tregs, in the irradiated tumor and in lymphoid organs. With the new PET tracer, we visualized the whole-body distribution of IL-2c and the bound receptors in naïve mice and tumor-bearing mice. Surprisingly, the tumor uptake was non-specific and only moderate. This prompted experiments demonstrating that specific IL-2c binding in the tumor is limited by IL-2 secreted by tumor-resident effector cells and that extratumorally expanded T and NK cells can infiltrate the irradiated tumor, which suggests that systemic immune activation considerably contributed to the reduction of tumor growth. Lastly, we show that a side effect of IL-2c treatment - a quite dramatic non-specific expansion of CD8+ T and NK cells - is only transient, and we visualized the associated splenomegaly as well as side effects on liver and lung by contrast-enhanced CT and PD-L1 PET. CONCLUSIONS: Our results show that the combination of immunogenic RT with IL-2c that are directed towards the low-affinity IL-2 receptor can be synergistic and more effective than the combination with uncomplexed IL-2. In addition, our theranostic evaluation provided insights into the mechanism of action and the side effects of IL-2c treatment.


Antibodies, Monoclonal/pharmacology , Interleukin-2/pharmacology , Neoplasms/pathology , Neoplasms/therapy , Radiation Dose Hypofractionation , Adoptive Transfer , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Biomarkers , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Drug Synergism , Humans , Immunologic Factors/pharmacology , Immunophenotyping , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Neoplasms/diagnostic imaging , Neoplasms/etiology , Positron Emission Tomography Computed Tomography , Positron-Emission Tomography , Species Specificity , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Xenograft Model Antitumor Assays
4.
Sci Transl Med ; 10(429)2018 02 21.
Article En | MEDLINE | ID: mdl-29467301

Recent evidence has revealed that oncogenic mutations may confer immune escape. A better understanding of how an oncogenic mutation affects immunosuppressive programmed death ligand 1 (PD-L1) expression may help in developing new therapeutic strategies. We show that oncogenic JAK2 (Janus kinase 2) activity caused STAT3 (signal transducer and activator of transcription 3) and STAT5 phosphorylation, which enhanced PD-L1 promoter activity and PD-L1 protein expression in JAK2V617F-mutant cells, whereas blockade of JAK2 reduced PD-L1 expression in myeloid JAK2V617F-mutant cells. PD-L1 expression was higher on primary cells isolated from patients with JAK2V617F-myeloproliferative neoplasms (MPNs) compared to healthy individuals and declined upon JAK2 inhibition. JAK2V617F mutational burden, pSTAT3, and PD-L1 expression were highest in primary MPN patient-derived monocytes, megakaryocytes, and platelets. PD-1 (programmed death receptor 1) inhibition prolonged survival in human MPN xenograft and primary murine MPN models. This effect was dependent on T cells. Mechanistically, PD-L1 surface expression in JAK2V617F-mutant cells affected metabolism and cell cycle progression of T cells. In summary, we report that in MPN, constitutive JAK2/STAT3/STAT5 activation, mainly in monocytes, megakaryocytes, and platelets, caused PD-L1-mediated immune escape by reducing T cell activation, metabolic activity, and cell cycle progression. The susceptibility of JAK2V617F-mutant MPN to PD-1 targeting paves the way for immunomodulatory approaches relying on PD-1 inhibition.


B7-H1 Antigen/metabolism , Hematologic Neoplasms/metabolism , Janus Kinase 2/metabolism , Myeloproliferative Disorders/metabolism , Animals , B7-H1 Antigen/genetics , Cell Proliferation/genetics , Cell Proliferation/physiology , Cell Transformation, Neoplastic , Hematologic Neoplasms/genetics , Humans , Janus Kinase 2/genetics , K562 Cells , Mice , Myeloproliferative Disorders/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Tumor Cells, Cultured
5.
Theranostics ; 6(10): 1629-40, 2016.
Article En | MEDLINE | ID: mdl-27446497

Checkpoint-blocking antibodies like those targeting the PD-1/PD-L1 pathway have revolutionized oncology. We developed radiotracers based on therapeutic checkpoint-blocking antibodies permitting sensitive and high-resolution PET imaging of both PD-1 and PD-L1 in immunocompetent mice. ImmunoPET of naive mice revealed similar overall expression patterns for PD-1 and PD-L1 in secondary lymphoid organs (spleen and lymph nodes). Interestingly, PD-L1 was also detected in brown adipose tissue (BAT), confirming the notion that BAT is immunologically relevant. Under pathophysiological conditions, strong expression of the receptor/ligand pair was also found in non-lymphoid tissues. Both were specifically detected in malignant tumors. PD-1 was readily detected after combined immunoradiotherapy causing massive tumor infiltration by PD-1+ lymphocytes. PD-L1 tracer uptake was reduced in PD-L1 knockout tumors. Moreover, monitoring the expression changes of PD-L1 in response to its main inducer, the effector T cell cytokine IFN-γ, revealed robust upregulation in the lung. This suggests that T cell responses in the lung, a vital organ continuously exposed to a variety of antigens, are strongly restrained by the PD-1 checkpoint. In turn, this could explain the association of PD-1 checkpoint inhibition with potentially fatal immune-mediated pneumonitis and partially also its efficacy in lung cancer.


Antibodies/administration & dosage , B7-H1 Antigen/analysis , Immunologic Factors/administration & dosage , Positron-Emission Tomography/methods , Programmed Cell Death 1 Receptor/analysis , Animals , B7-H1 Antigen/immunology , Mice , Programmed Cell Death 1 Receptor/immunology
6.
Cancer Res ; 76(16): 4673-83, 2016 08 15.
Article En | MEDLINE | ID: mdl-27302161

T cell-recruiting bispecific antibodies (bsAb) show promise in hematologic malignancies and are also being evaluated in solid tumors. In this study, we investigated whether T cell-recruiting bsAbs synergize with hypofractionated tumor radiotherapy (hRT) and/or blockade of the programmed death-1 (PD-1) immune checkpoint, both of which can increase tumor-infiltrating lymphocyte (TIL) numbers. Unexpectedly, large melanomas treated with hRT plus bsAb (AC133×CD3) relapsed faster than those treated with hRT alone, accompanied by massive TIL apoptosis. This fast relapse was delayed by the further addition of anti-PD-1. Mechanistic investigations revealed restimulation-induced cell death mediated by BIM and FAS as an additional cause of bsAb-mediated TIL depletion. In contrast, the double combination of hRT and anti-PD-1 strongly increased TIL numbers, and even very large tumors were completely eradicated. Our study reveals the risk that CD3-engaging bsAbs can induce apoptotic TIL depletion followed by rapid tumor regrowth, reminiscent of tolerance induction by CD3 mAb-mediated T-cell depletion, warranting caution in their use for the treatment of solid tumors. Our findings also argue that combining radiotherapy and anti-PD-1 can be quite potent, including against very large tumors. Cancer Res; 76(16); 4673-83. ©2016 AACR.


Antibodies, Bispecific/administration & dosage , Antineoplastic Agents/administration & dosage , CD8-Positive T-Lymphocytes/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Melanoma, Experimental/pathology , Animals , Antibodies, Bispecific/adverse effects , Antineoplastic Agents/adverse effects , CD8-Positive T-Lymphocytes/radiation effects , Chemoradiotherapy/methods , Flow Cytometry , Lymphocytes, Tumor-Infiltrating/radiation effects , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Radiotherapy
7.
Cancer Res ; 75(11): 2166-76, 2015 Jun 01.
Article En | MEDLINE | ID: mdl-25840983

Cancer stem cells (CSC) drive tumorigenesis and contribute to genotoxic therapy resistance, diffuse infiltrative invasion, and immunosuppression, which are key factors for the incurability of glioblastoma multiforme (GBM). The AC133 epitope of CD133 is an important CSC marker for GBM and other tumor entities. Here, we report the development and preclinical evaluation of a recombinant AC133×CD3 bispecific antibody (bsAb) that redirects human polyclonal T cells to AC133(+) GBM stem cells (GBM-SC), inducing their strong targeted lysis. This novel bsAb prevented the outgrowth of AC133-positive subcutaneous GBM xenografts. Moreover, upon intracerebral infusion along with the local application of human CD8(+) T cells, it exhibited potent activity in prophylactic and treatment models of orthotopic GBM-SC-derived invasive brain tumors. In contrast, normal hematopoietic stem cells, some of which are AC133-positive, were virtually unaffected at bsAb concentrations effective against GBM-SCs and retained their colony-forming abilities. In conclusion, our data demonstrate the high activity of this new bsAb against patient-derived AC133-positive GBM-SCs in models of local therapy of highly invasive GBM.


Antibodies, Bispecific/therapeutic use , Antigens, CD/immunology , Glioblastoma/therapy , Glycoproteins/immunology , Neoplastic Stem Cells/immunology , Peptides/immunology , AC133 Antigen , Antibodies, Bispecific/immunology , Antigens, CD/therapeutic use , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Carcinogenesis/immunology , Cell- and Tissue-Based Therapy/methods , Epitopes/immunology , Glioblastoma/immunology , Glioblastoma/pathology , Glycoproteins/therapeutic use , Humans , Immunotherapy/methods , Neoplastic Stem Cells/pathology , Peptides/therapeutic use
8.
Oncotarget ; 6(1): 171-84, 2015 Jan 01.
Article En | MEDLINE | ID: mdl-25426558

The AC133 epitope of CD133 is a cancer stem cell (CSC) marker for many tumor entities, including the highly malignant glioblastoma multiforme (GBM). We have developed an AC133-specific chimeric antigen receptor (CAR) and show that AC133-CAR T cells kill AC133+ GBM stem cells (GBM-SCs) both in vitro and in an orthotopic tumor model in vivo. Direct contact with patient-derived GBM-SCs caused rapid upregulation of CD57 on the CAR T cells, a molecule known to mark terminally or near-terminally differentiated T cells. However, other changes associated with terminal T cell differentiation could not be readily detected. CD57 is also expressed on tumor cells of neural crest origin and has been preferentially found on highly aggressive, undifferentiated, multipotent CSC-like cells. We found that CD57 was upregulated on activated T cells only upon contact with CD57+ patient-derived GBM-SCs, but not with conventional CD57-negative glioma lines. However, CD57 was not downregulated on the GBM-SCs upon their differentiation, indicating that this molecule is not a bona fide CSC marker for GBM. Differentiated GBM cells still induced CD57 on CAR T cells and other activated T cells. Therefore, CD57 can apparently be upregulated on activated human T cells by mere contact with CD57+ target cells.


Antigens, CD/metabolism , Brain Neoplasms/metabolism , CD57 Antigens/metabolism , Glioblastoma/metabolism , Glycoproteins/metabolism , Peptides/metabolism , AC133 Antigen , Animals , Brain Neoplasms/pathology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Flow Cytometry , Glioblastoma/pathology , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/cytology , Receptors, Antigen/metabolism , T-Lymphocytes/immunology , Up-Regulation
9.
Proc Natl Acad Sci U S A ; 111(6): E692-701, 2014 Feb 11.
Article En | MEDLINE | ID: mdl-24469819

A technology that visualizes tumor stem cells with clinically relevant tracers could have a broad impact on cancer diagnosis and treatment. The AC133 epitope of CD133 currently is one of the best-characterized tumor stem cell markers for many intra- and extracranial tumor entities. Here we demonstrate the successful noninvasive detection of AC133(+) tumor stem cells by PET and near-infrared fluorescence molecular tomography in subcutaneous and orthotopic glioma xenografts using antibody-based tracers. Particularly, microPET with (64)Cu-NOTA-AC133 mAb yielded high-quality images with outstanding tumor-to-background contrast, clearly delineating subcutaneous tumor stem cell-derived xenografts from surrounding tissues. Intracerebral tumors as small as 2-3 mm also were clearly discernible, and the microPET images reflected the invasive growth pattern of orthotopic cancer stem cell-derived tumors with low density of AC133(+) cells. These data provide a basis for further preclinical and clinical use of the developed tracers for high-sensitivity and high-resolution monitoring of AC133(+) tumor stem cells.


Antigens, CD/immunology , Glycoproteins/immunology , Neoplastic Stem Cells/immunology , Peptides/immunology , Positron-Emission Tomography/methods , AC133 Antigen , Animals , Brain Neoplasms/diagnostic imaging , Fluorescence , Glioblastoma/diagnostic imaging , Heterografts , Mice , Multimodal Imaging , Tomography, X-Ray Computed
10.
J Affect Disord ; 87(2-3): 305-11, 2005 Aug.
Article En | MEDLINE | ID: mdl-15951024

BACKGROUND: Recent studies showed an activation of the cytokine system and the HPA axis in major depression, although with inconsistent results. While the non-melancholic subtype displayed a proinflammatory cytokine pattern, the melancholic subtype showed signs of impaired cytokine production. In order to understand the potential pathogenic significance of these systems further, the interplay between the cytokine system and the HPA axis in depressive subtypes as well as potential changes of these systems during the course of disease were investigated. METHODS: N=37 initially unmedicated patients with acute major depression were sub-classified (melancholic vs. non-melancholic) and compared with N=37 matched healthy controls. Upon admission and after complete clinical remission, basal plasma ACTH and serum cortisol levels as well as cytokine productions (IL-1beta, IL-1 receptor antagonist (IL-1RA)) upon mitogen stimulation (PHA) were measured in a whole blood assay. RESULTS: ACTH and cortisol concentrations were significantly elevated on admission in the melancholic but not the non-melancholic subgroup. Non-melancholic patients produced significantly more IL-1beta and IL-1RA upon admission than controls or melancholic patients. The IL-1 RA/IL-1beta ratio was significantly lower in the non-melancholic compared to the melancholic subgroup and increased significantly upon remission. LIMITATIONS: Patient treatment was not standardized. No Dex/CRH test was performed. CONCLUSIONS: Melancholic patients demonstrated an activation of the HPA axis in acute stage with partial normalization upon remission but no signs of inflammation. Non-melancholic patients showed signs of inflammation in acute depression with normalization upon remission while the function of the HPA axis was normal.


Cytokines/immunology , Depressive Disorder, Major/immunology , Depressive Disorder, Major/physiopathology , Hypothalamo-Hypophyseal System/physiopathology , Pituitary-Adrenal System/physiopathology , Adrenocorticotropic Hormone/blood , Cytokines/blood , Depressive Disorder, Major/blood , Depressive Disorder, Major/diagnosis , Diagnostic and Statistical Manual of Mental Disorders , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hydrocortisone/blood , Interleukin-1/blood , Interleukin-1/immunology , Male , Middle Aged , Receptors, Interleukin-1/blood , Receptors, Interleukin-1/immunology
11.
Psychiatr Prax ; 30(2): 83-8, 2003 Mar.
Article De | MEDLINE | ID: mdl-12601597

OBJECTIVE: The work of a psychiatric department (in Rhede/Westphalia) was statistically investigated by compiling the most important patient and therapy data in five year periods over its first decade of existence. METHODS: Data of 1364 admissions were surveyed in the years 1983, 1988 and 1993. These data were statistically analysed and showed age, sex, social class, living conditions, place of residence, distance to hospital, diagnoses, duration of stay, readmission, pre- and aftercare. RESULTS: The number of patients living in the catchmentarea increased, the duration of stay decreased, readmissions increased, the rate of patients without aftercare decreased, the rate of diagnoses kept continuously, patients were more often admitted to hospital directly. CONCLUSIONS: These and further results point out an increasing extent of community based psychiatry in the region.


Mental Disorders/epidemiology , Patient Admission/statistics & numerical data , Psychiatric Department, Hospital/statistics & numerical data , Socioeconomic Factors , Adult , Aftercare/statistics & numerical data , Aged , Catchment Area, Health/statistics & numerical data , Community Mental Health Services/statistics & numerical data , Cross-Sectional Studies , Female , Germany/epidemiology , Health Services Accessibility/statistics & numerical data , Humans , Length of Stay/statistics & numerical data , Male , Mental Disorders/diagnosis , Mental Disorders/therapy , Middle Aged , Patient Readmission/statistics & numerical data
...