Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Leuk Res ; 117: 106842, 2022 06.
Article in English | MEDLINE | ID: mdl-35490594

ABSTRACT

Cancer testis antigen PRAME is over-expressed in a variety of malignant cells but is not or minimally expressed in normal non-germ line cells. Adoptive transfer of PRAME-specific T cells is thus under investigation in clinical trials as an innovative therapeutic option for acute myeloid leukemia (AML). However, their senescence-inducing activity has not been studied. This study therefore examines senescence induction in AML cells by PRAME-specific TH1 cells. Analysis of cell cycle and marker expression demonstrate that the supernatants of antigen-stimulated PRAME-specific TH1 cells induce senescence in AML cell lines Kasumi and Nomo-1 through combinative IFN-γ and TNF-α. Additionally IFN-γ and TNF-α secreted by TCR-activated Vδ2+ or CMV-specific T cells can also drive these AML cell lines into terminal growth arrest. G1/0 arrest is also suggested in patient-derived AML by TH1 cytokines or supernatants from Zoledronate-stimulated or aCD3/aCD28-stimulated PBMCs. Thus, we show for the first time that senescence is induced in AML cells by combined IFN-γ and TNF-α, and that these cytokines can be derived either from TCR-engineered CD4+ T cells, or intriguingly from Virus-specific as well as innate Vδ2+ T cells responding to their cognate antigens, namely T-cell responses targeting an antigen that is NOT expressed by the leukemic cells.


Subject(s)
Cytokines , Leukemia, Myeloid, Acute , Humans , Interferon-gamma/metabolism , Leukemia, Myeloid, Acute/therapy , Male , Receptors, Antigen, T-Cell , Tumor Necrosis Factor-alpha
2.
Bone Marrow Transplant ; 56(10): 2336-2354, 2021 10.
Article in English | MEDLINE | ID: mdl-33976380

ABSTRACT

Detrimental graft-versus-host disease (GVHD) still remains a major cause of death in hematopoietic stem cell transplantation (HSCT). The recently explored depletion of naive cells from mobilized grafts (CD45RA depletion) has shown considerable promise, yet is unable to eliminate the incidence of GVHD. Analysis of CD45RA-depleted haploidentical mixed lymphocytes culture (haplo-MLC) revealed insufficient suppression of alloresponses in the CD4+ compartment and identified CD276 as a marker for alloreactive memory Th1 T cells. Conclusively, depleting CD276+ cells from CD45RA-depleted haplo-MLC significantly attenuated alloreactivity to recipient cells while increasing antiviral reactivity and maintaining anti-third party reactivity in vitro. To evaluate these findings in vivo, bulk, CD45RA-depleted, or CD45RA/CD276-depleted CD4+ T cells from HLA-DR4negative healthy humans were transplanted into NSG-Ab°DR4 mice, a sensitive human allo-GVHD model. Compellingly, CD45RA/CD276-depleted grafts from HLA-DR4negative donors or in vivo depletion of CD276+ cells after transplant of HLA-DR4negative memory CD4 T cells significantly delay the onset of GVHD symptoms and significantly alleviate its severity in NSG-Ab°DR4 mice. The clinical courses correlated with diminished Th1-cytokine secretion and downregulated CXCR6 expression of engrafted peripheral T cells. Collectively, mismatched HLA-mediated GVHD can be controlled by depleting recipient-specific CD276+ alloreacting T cells from the graft, highlighting its application in haplo-HSCT.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Animals , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Immunologic Memory , Lymphocyte Depletion , Mice , T-Lymphocytes
3.
Oncoimmunology ; 10(1): 2003532, 2021.
Article in English | MEDLINE | ID: mdl-35686214

ABSTRACT

Chimeric antigen receptor (CAR)-T therapy holds great promise to sustainably improve cancer treatment. However, currently, a broad applicability of CAR-T cell therapies is hampered by limited CAR-T cell versatility and tractability and the lack of exclusive target antigens to discriminate cancerous from healthy tissues. To achieve temporal and qualitative control on CAR-T function, we engineered the Adapter CAR (AdCAR) system. AdCAR-T are redirected to surface antigens via biotin-labeled adapter molecules in the context of a specific linker structure, referred to as Linker-Label-Epitope. AdCAR-T execute highly specific and controllable effector function against a multiplicity of target antigens. In mice, AdCAR-T durably eliminate aggressive lymphoma. Importantly, AdCAR-T might prevent antigen evasion by combinatorial simultaneous or sequential targeting of multiple antigens and are capable to identify and differentially lyse cancer cells by integration of adapter molecule-mediated signals based on multiplex antigen expression profiles. In consequence the AdCAR technology enables controllable, flexible, combinatorial, and selective targeting.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Animals , Immunotherapy, Adoptive , Mice , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocytes , Technology
4.
Int J Oncol ; 57(1): 289-300, 2020 07.
Article in English | MEDLINE | ID: mdl-32377699

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common type of pediatric soft tissue sarcoma. The prognosis of advanced stage RMS remains poor, and metastatic invasion is a major cause of treatment failure. Therefore, there is an urgent need for treatment alternatives focusing on metastatic invasion and drug resistance. The stromal cell­derived factor­1 (SDF­1)/chemokine receptor 4 (CXCR4) axis is a crucial factor for metastatic invasion in RMS. Clinical data has revealed that high CXCR4 expression is associated with a poor outcome and a high metastatic rate in several malignancies, including RMS. Thus, targeting CXCR4 in addition to classical chemotherapy may improve the effectiveness of RMS treatment. In the present study, flow cytometry and reverse transcription­quantitative PCR were used to assess the effects of the combined treatment with a CXCR4 antagonist and chemotherapy on CXCR4 expression in the embryonal RMS (RME) cell line RD and in the alveolar RMS (RMA) cell line RH30. The functional effect of CXCR4 expression on the migratory behavior of RMS cells was analyzed using Transwell assays. Treatment with cytotoxic agents modulated CXCR4 expression in RMS cells in a dose­, drug­ and cell line dependent manner; however, this was not observed in RD cells with vincristine. The expression levels of CXCR4 significantly increased the migratory behavior of RMA and did not affect RME cell migration towards stromal cell­derived factor­1α (SDF­1α). AMD3100 markedly reduced the migration of RH30 cells in the Transwell assays compared with SDF­1α alone, and the cytotoxic agents doxorubicin and vincristine increased this effect. The results of the combined treatment in RMS cells using the CXCR4 antagonist AMD3100 together with cytotoxic drugs demonstrated that this approach may be a promising alternative for the treatment of advanced stage pediatric RMS. The observed effects of circumventing metastatic invasion and drug resistance should be further investigated in vivo.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzylamines/pharmacology , Cyclams/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Rhabdomyosarcoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzylamines/therapeutic use , Cell Line, Tumor , Cell Movement , Chemokine CXCL12/analysis , Chemokine CXCL12/metabolism , Child, Preschool , Cyclams/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Infant , Male , Prospective Studies , Receptors, CXCR4/analysis , Receptors, CXCR4/metabolism , Rhabdomyosarcoma/pathology , Signal Transduction/drug effects , Vincristine/pharmacology , Vincristine/therapeutic use
5.
Oncoimmunology ; 9(1): 1683345, 2020.
Article in English | MEDLINE | ID: mdl-32002293

ABSTRACT

Expression of the disialoganglioside GD2 has been identified as a marker antigen associated with a breast cancer stem-like cell (BCSC) phenotype. Here, we report on the evaluation of GD2 as a BCSC-specific target antigen for immunotherapy. GD2 expression was confirmed at variable degree in a set of breast cancer cell lines, predominantly in triple-negative breast cancer (TNBC). To target GD2, we have generated novel anti-GD2 chimeric antigen receptors (GD2-CAR), based on single-chain variable fragments (scFv) derived from the monoclonal antibody (mAb) ch14.18, also known as dinutuximab beta. Expressed on T cells, GD2-CARs mediated specific GD2-dependent T-cell activation and target cell lysis. In contrast to previously described GD2-CARs, no signs of exhaustion by tonic signaling were found. Importantly, application of GD2-CAR expressing T cells (GD2-CAR-T) in an orthotopic xenograft model of TNBC (MDA-MB-231) halted local tumor progression and completely prevented lung metastasis formation. In line with the BCSC model, GD2 expression was only found in a subpopulation (4-6%) of MDA-MB-231 cells before injection. Significant expansion of GD2-CAR-T in tumor-bearing mice as well as T-cell infiltrates in the primary tumor and the lungs were found, indicating site-specific activation of GD2-CAR-T. Our data strongly support previous findings of GD2 as a BCSC-associated antigen. GD2-targeted immunotherapies have been extensively studied in human. In conclusion, GD2-CAR-T should be considered a promising novel approach for GD2-positive breast cancer, especially to eliminate disseminated tumor cells and prevent metastasis formation.


Subject(s)
Breast Neoplasms , Immunotherapy, Adoptive , Neoplasm Metastasis/prevention & control , Neoplastic Stem Cells , Receptors, Chimeric Antigen , Animals , Breast Neoplasms/therapy , Female , Gangliosides , Humans , Mice , Receptors, Chimeric Antigen/genetics , T-Lymphocytes
6.
Cancer Immunol Immunother ; 69(4): 593-610, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31982940

ABSTRACT

Despite recent progress in the understanding of γδ T cells' roles and functions, their interaction with αß T cells still remains to be elucidated. In this study, we sought to clarify what precisely endows peripheral Vδ2+ T cells with immunosuppressive function on autologous αß T cells. We found that negatively freshly isolated Vδ2+ T cells do not exhibit suppressive behavior, even after stimulation with IL-12/IL-18/IL-15 or the sheer contact with butyrophilin-3A1-expressing tumor cell lines (U251 or SK-Mel-28). On the other hand, Vδ2+ T cells positively isolated through TCR crosslinking or after prolonged stimulation with isopentenyl pyrophosphate (IPP) mediate strong inhibitory effects on αß T cell proliferation. Stimulation with IPP in the presence of IL-15 induces the most robust suppressive phenotype of Vδ2+ T cells. This indicates that Vδ2+ T cells' suppressive activity is dependent on a TCR signal and that the degree of suppression correlates with its strength. Vδ2+ T cell immunosuppression does not correlate with their Foxp3 expression but rather with their PD-L1 protein expression, evidenced by the massive reduction of suppressive activity when using a blocking antibody. In conclusion, pharmacologic stimulation of Vδ2+ T cells via the Vδ2 TCR for activation and expansion induces Vδ2+ T cells' potent killer activity while simultaneously licensing them to suppress αß T cell responses. Taken together, the study is a further step to understand-in more detail-the suppressive activity of Vδ2+ γδ T cells.


Subject(s)
Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Apoptosis/drug effects , Apoptosis/immunology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Gene Expression/drug effects , Gene Expression/immunology , Hemiterpenes/pharmacology , Humans , Immune Tolerance/drug effects , Immune Tolerance/genetics , Immune Tolerance/immunology , Interleukin-15/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Organophosphorus Compounds/pharmacology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Signal Transduction/drug effects , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/metabolism
7.
J Leukoc Biol ; 107(6): 1097-1105, 2020 06.
Article in English | MEDLINE | ID: mdl-31967358

ABSTRACT

Dissection of the role and function of human γδ T cells and their heterogeneous subsets in cancer, inflammation, and auto-immune diseases is a growing and dynamic research field of increasing interest to the scientific community. Therefore, harmonization and standardization of techniques for the characterization of peripheral and tissue-resident γδ T cells is crucial to facilitate comparability between published and emerging research. The application of commercially available reagents to classify γδ T cells, in particular the combination of multiple Abs, is not always trouble-free, posing major demands on researchers entering this field. Occasionally, even entire γδ T cell subsets may remain undetected when certain Abs are combined in flow cytometric analysis with multicolor Ab panels, or might be lost during cell isolation procedures. Here, based on the recent literature and our own experience, we provide an overview of methods commonly employed for the phenotypic and functional characterization of human γδ T cells including advanced polychromatic flow cytometry, mass cytometry, immunohistochemistry, and magnetic cell isolation. We highlight potential pitfalls and discuss how to circumvent these obstacles.


Subject(s)
Flow Cytometry/standards , Immunomagnetic Separation/standards , Receptors, Antigen, T-Cell, gamma-delta/analysis , T-Lymphocyte Subsets/immunology , Antibodies/chemistry , Carcinoma/diagnosis , Carcinoma/immunology , Carcinoma/pathology , Case-Control Studies , Colonic Neoplasms/diagnosis , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Flow Cytometry/methods , Fluorescent Dyes/chemistry , Gene Expression , Hodgkin Disease/diagnosis , Hodgkin Disease/immunology , Hodgkin Disease/pathology , Humans , Immunohistochemistry , Immunomagnetic Separation/methods , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/pathology
8.
Cancers (Basel) ; 12(1)2020 Jan 04.
Article in English | MEDLINE | ID: mdl-31947966

ABSTRACT

Abundant IFN-γ secretion, potent cytotoxicity, and major histocompatibility complex-independent targeting of a large spectrum of tumors make γδ T cells attractive candidates for cancer immunotherapy. Upon tumor recognition through the T-cell receptor (TCR), NK-receptors, or NKG2D, γδ T cells generate the pro-inflammatory cytokines TNF-α and IFN-γ, or granzymes and perforin that mediate cellular apoptosis. Despite these favorable potentials, most clinical trials testing the adoptive transfer of pharmacologically TCR-targeted and expanded γδ T cells resulted in a limited response. Recently, the TCR-independent activation of γδ T cells was identified. However, the modulation of γδ T cell's effector functions solely by cytokines remains to be elucidated. In the present study, we systematically analyzed the impact of IL-2, IL-12, and IL-18 in parallel with TCR stimulation on proliferation, cytokine production, and anti-tumor activity of γδ T cells. Our results demonstrate that IL-12 and IL-18, when combined, constitute the most potent stimulus to enhance anti-tumor activity and induce proliferation and IFN-γ production by γδ T cells in the absence of TCR signaling. Intriguingly, stimulation with IL-12 and IL-18 without TCR stimulus induces a comparable degree of anti-tumor activity in γδ T cells to TCR crosslinking by killing tumor cells and driving cancer cells into senescence. These findings approve the use of IL-12/IL-18-stimulated γδ T cells for adoptive cell therapy to boost anti-tumor activity by γδ T cells.

9.
BMC Pediatr ; 19(1): 470, 2019 Dec 02.
Article in English | MEDLINE | ID: mdl-31791281

ABSTRACT

After publication of the original article (1), it was brought to our attention that references 24 and 31 are inappropriately cited in the article.

10.
BMC Pediatr ; 19(1): 346, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31604460

ABSTRACT

BACKGROUND: DNA ligase IV deficiency is a rare autosomal recessive disorder caused by hypomorphic mutations in the DNA ligase IV (LIG4) gene. DNA ligase IV is an essential protein for the development of a healthy immune system as well as for the protection of genomic integrity. Apart from typical stigmata, patients with DNA ligase IV deficiency are characterized by progressive bone marrow failure and a predisposition to malignancy. To our knowledge this reported case is the first description of two brothers with ligase IV deficiency who are treated with different hematopoietic stem cell transplantation (HSCT) regimens resulting in vastly divergent outcomes. CASE PRESENTATION: The cases of two brothers suffering from severe recurrent infections and growth retardation are described. The laboratory findings showed pancytopenia with significant lymphopenia. The two boys were diagnosed with DNA ligase IV deficiency, associated with severe combined immunodeficiency (SCID). Both patients received HSCT from two different matched unrelated donors (MUD) at the age of 33 and 18 months. The older brother succumbed post-transplant due to fatal side-effects 143 days after allogeneic HSCT. The younger brother - conditioned with a different regimen - received a T cell depleted graft 4 months later. No severe side-effects occurred, neither post-transplant nor in the following years. Ten years after HSCT the patient is well off, living a normal life and attending a regular high school. His immune system is fully reconstituted, resulting in a maximum of T cell receptor (TCR) diversity, which is a prerequisite for immune competence. However, he still suffers from microcephaly, dwarfism and dystrophy. CONCLUSIONS: This case report gives an example of a successful HSCT as a treatment option in a genetic disorder such as ligase IV deficiency, using a rather mild conditioning regimen. Further studies are required to determine the viability and efficacy of this treatment option.


Subject(s)
DNA Ligase ATP/deficiency , Hematopoietic Stem Cell Transplantation/methods , Severe Combined Immunodeficiency/complications , Siblings , Transplantation Conditioning/methods , Child, Preschool , Fatal Outcome , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Infant , Male , Severe Combined Immunodeficiency/immunology , Transplantation, Homologous
11.
J Transl Med ; 16(1): 23, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29409514

ABSTRACT

BACKGROUND: Cancer vaccines can effectively establish clinically relevant tumor immunity. Novel sequencing approaches rapidly identify the mutational fingerprint of tumors, thus allowing to generate personalized tumor vaccines within a few weeks from diagnosis. Here, we report the case of a 62-year-old patient receiving a four-peptide-vaccine targeting the two sole mutations of his pancreatic tumor, identified via exome sequencing. METHODS: Vaccination started during chemotherapy in second complete remission and continued monthly thereafter. We tracked IFN-γ+ T cell responses against vaccine peptides in peripheral blood after 12, 17 and 34 vaccinations by analyzing T-cell receptor (TCR) repertoire diversity and epitope-binding regions of peptide-reactive T-cell lines and clones. By restricting analysis to sorted IFN-γ-producing T cells we could assure epitope-specificity, functionality, and TH1 polarization. RESULTS: A peptide-specific T-cell response against three of the four vaccine peptides could be detected sequentially. Molecular TCR analysis revealed a broad vaccine-reactive TCR repertoire with clones of discernible specificity. Four identical or convergent TCR sequences could be identified at more than one time-point, indicating timely persistence of vaccine-reactive T cells. One dominant TCR expressing a dual TCRVα chain could be found in three T-cell clones. The observed T-cell responses possibly contributed to clinical outcome: The patient is alive 6 years after initial diagnosis and in complete remission for 4 years now. CONCLUSIONS: Therapeutic vaccination with a neoantigen-derived four-peptide vaccine resulted in a diverse and long-lasting immune response against these targets which was associated with prolonged clinical remission. These data warrant confirmation in a larger proof-of concept clinical trial.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Carcinoma, Pancreatic Ductal/therapy , Epitopes/immunology , Monitoring, Immunologic , Pancreatic Neoplasms/therapy , Receptors, Antigen, T-Cell, alpha-beta/genetics , Vaccines, Subunit/immunology , Amino Acid Sequence , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/secondary , Humans , Male , Middle Aged , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/secondary , Peptides/chemistry , Peptides/immunology , Treatment Outcome , Vaccination
12.
Blood ; 131(10): 1063-1072, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29358176

ABSTRACT

Allogeneic hematopoetic stem cell transplantation (HCT) offers an option for patients with hematologic malignancies, in whom conventional standard therapies failed or are not effective enough to cure the disease. Successful HCT can restore functional hematopoiesis and immune function, and the new donor-derived immune system can exert a graft-versus-leukemia (GVL) effect. However, allogenic HCT can also be associated with serious risks for transplantation-related morbidities or mortalities such as graft-versus-host disease (GVHD) or life-threatening infectious complications. GVHD is caused by alloreactive T lymphocytes, which express the αß T-cell receptor, whereas lymphocytes expressing the γδ T-cell receptor are not alloreactive and do not induce GVHD but can exhibit potent antileukemia and anti-infectious activities. Therefore, γδ T cells are becoming increasingly interesting in allogeneic HCT, and clinical strategies to exploit the full function of these lymphocytes have been and are being developed. Such strategies comprise the in vivo activation of γδ T cells or subsets after HCT by certain drugs or antibodies or the ex vivo expansion and manipulation of either patient-derived or donor-derived γδ T cells and their subsets and the adoptive transfer of the ex vivo-activated lymphocytes. On the basis of the absence of dysregulated alloreactivity, such approaches could induce potent GVL effects in the absence of GVHD. The introduction of large-scale clinical methods to enrich, isolate, expand, and manipulate γδ T cells will facilitate future clinical studies that aim to exploit the full function of these beneficial nonalloreactive lymphocytes.


Subject(s)
Graft vs Host Disease , Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cell Transplantation , Infections , Leukemia , Lymphocyte Transfusion , Receptors, Antigen, T-Cell, gamma-delta/immunology , Allografts , Animals , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Humans , Infections/immunology , Infections/pathology , Infections/therapy , Leukemia/pathology , Leukemia/therapy , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/transplantation , Tissue Donors
13.
Front Immunol ; 9: 3018, 2018.
Article in English | MEDLINE | ID: mdl-30622535

ABSTRACT

Cancer immunotherapy has been established as standard of care in different tumor entities. After the first reports on synergistic effects with radiotherapy and the induction of abscopal effects-tumor shrinkage outside the irradiated volume attributed to immunological effects of radiotherapy-several treatment combinations have been evaluated. Different immunotherapy strategies (e.g., immune checkpoint inhibition, vaccination, cytokine based therapies) have been combined with local tumor irradiation in preclinical models. Clinical trials are ongoing in different cancer entities with a broad range of immunotherapeutics and radiation schedules. SDF-1 (CXCL12)/CXCR4 signaling has been described to play a major role in tumor biology, especially in hypoxia adaptation, metastasis and migration. Local tumor irradiation is a known inducer of SDF-1 expression and release. CXCR4 also plays a major role in immunological processes. CXCR4 antagonists have been approved for the use of hematopoietic stem cell mobilization from the bone marrow. In addition, several groups reported an influence of the SDF-1/CXCR4 axis on intratumoral immune cell subsets and anti-tumor immune response. The aim of this review is to merge the knowledge on the role of SDF-1/CXCR4 in tumor biology, radiotherapy and immunotherapy of cancer and in combinatorial approaches.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Chemokine CXCL12/metabolism , Immunotherapy/methods , Neoplasms/therapy , Receptors, CXCR4/antagonists & inhibitors , Antineoplastic Agents, Immunological/pharmacology , Bone Marrow/drug effects , Chemokine CXCL12/immunology , Chemoradiotherapy/methods , Clinical Trials as Topic , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Gene Expression Regulation, Neoplastic/radiation effects , Hematopoietic Stem Cell Mobilization/methods , Humans , Neoplasms/genetics , Neoplasms/immunology , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Treatment Outcome
14.
Immunol Res ; 66(1): 79-86, 2018 02.
Article in English | MEDLINE | ID: mdl-29138980

ABSTRACT

Increased numbers of CD8+CD28neg. T cells have been detected in the peripheral blood of patients with several types of malignancies. However, the role of these cells in anticancer immunity are not yet clear and CD8+CD28neg. T cells are a controversially discussed subpopulation reported both as immunosuppressive and cytotoxic. In this study, we examined the T cell receptor (TCR) repertoire and complementarity-determining region 3 sequences of CD28neg. T cells in a melanoma patient with recurrent disease who achieved long-term disease-free status. As a result, the patient's oligoclonal CD8+CD28neg. T cell compartment holds TCRs that are public and specific for Melan-A as well as several public TCRs reported for common viral antigens. While over 80% of his CD8+CD28neg. T cells expressed a cytotoxicity marker, CD57, only 0.01% of CD8+ CD28neg. T cells were positive for Foxp3. In conclusion, our results demonstrate that besides virus-specific also tumor-associated self-antigen targeting T cells accumulate in the CD28neg. compartment of the immunological memory. Since the patient is in ongoing complete remission for more than 9 years, CD8+CD28neg. T cells with the Melan-A-specific TCR might contribute to antitumor immunity in this patient.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Complementarity Determining Regions/genetics , Ear Neoplasms/immunology , Melanoma/immunology , Receptors, Antigen, T-Cell/genetics , Adult , CD28 Antigens/metabolism , Cytotoxicity, Immunologic , Ear Neoplasms/surgery , Humans , Immunity, Cellular , Immunologic Memory , MART-1 Antigen/immunology , Male , Margins of Excision , Melanoma/surgery , Recurrence , Remission Induction , T-Cell Antigen Receptor Specificity
15.
Oncoimmunology ; 6(6): e1323161, 2017.
Article in English | MEDLINE | ID: mdl-28680762

ABSTRACT

NHS-IL12 is an immunocytokine, a fusion protein of IL12's functional domains and a necrosis-targeting antibody, which has shown significant effects against human rhabdomyosarcoma xenografts in a humanized tumor model, including terminal growth arrest and differentiation of the tumor cells. Here, we locally irradiated the tumors, increasing necrosis and consequently intratumoral immune cytokine availability, and asked whether this effect may surmount efficacy of single treatment modality. Humanized mice bearing bilateral rhabdomyosarcoma xenografts were evaluated for tumor burden and survival after irradiation, systemic NHS-IL12 therapy or a combination of both. Intratumoral immune compartments were characterized by immunohistochemistry and molecular methods. TH1-cytokine dependency of underlying effector mechanisms were investigated in vitro in several human tumor cell lines. NHS-IL12 when combined with irradiation terminally arrested tumor growth and significantly improved survival. Combination treatment induced dense intratumoral T-cell infiltrates, clonal epitope-specific T-cell expansions, expression of cytotoxins, decreased pro-tumorigenic cytokines and induced senescence and differentiation in the cancer cells. Senescence and differentiation were reproduced in vitro and confirmed to be dependent on TH1 cytokines IFNγ and TNF-α. NHS-IL12 and irradiation together induced broad intratumoral TH1 biased NK and T-cell compartments, established antitumoral cytokine profiles and irreversibly growth arrested tumor cells, leading to systemic cancer control and improved survival. For the first time, we describe immune-induced senescence as a novel mechanism resulting from a treatment regimen combining irradiation with immunotherapy.

16.
Cancer Immunol Immunother ; 65(8): 1003-13, 2016 08.
Article in English | MEDLINE | ID: mdl-27376889

ABSTRACT

PURPOSE: NHS-IL12 is an immunocytokine targeting necrotic tumour areas. IL12 shows anti-tumour activity. As local irradiation might induce additional necrosis in solid tumours, we aimed to evaluate the increase in intratumoural accumulation of NHS-IL12 after irradiation and correlate the findings with diffusion-weighted MRI studies in two xenograft models. METHODS: Human rhabdomyosarcoma (A204) and prostate cancer (PC3) cells were studied in vitro and as subcutaneous xenografts. Radiation sensitivity of the cell lines was assessed in vitro by colony formation assays. In vivo tumour necrosis was assessed based on apparent diffusion coefficients (ADC). Biodistribution of NHS-IL12 was evaluated with and without tumour irradiation using in vivo small-animal PET and ex vivo biodistribution. RESULTS: A204 and PC3 differed in their intrinsic radiation sensitivity. Accordingly, radiation-induced tumour necrosis was found only in A204 xenografts. In comparison with control, ADC was significantly increased after irradiation of A204 tumours with 1 × 8.0 Gy and 5 × 2.0 Gy, whereas no change in ADC was observed in PC3 xenografts in all irradiation regimes. ADC correlated with histology. An enhanced uptake of radiolabelled NHS-IL12 in A204 tumours was detected by PET and ex vivo biodistribution after tumour irradiation. In PC3 tumours, no increase in NHS-IL12 uptake was observed. CONCLUSIONS: In dependence of the tumour model, tumour irradiation enhanced tumour necrosis measured in MRI and histology. In vivo PET and ex vivo biodistribution showed enhanced binding of NHS-IL12 in rhabdomyosarcoma xenografts. Thus, enhanced binding of necrosis-targeting immunocytokines might be a novel mechanism of additive effects in combination with irradiation.


Subject(s)
Immunoconjugates/immunology , Prostatic Neoplasms/genetics , Rhabdomyosarcoma/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Male , Mice
17.
Oncotarget ; 7(12): 14259-78, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26893360

ABSTRACT

Infiltration of the brain by glioblastoma cells reportedly requires Ca2+ signals and BK K+ channels that program and drive glioblastoma cell migration, respectively. Ionizing radiation (IR) has been shown to induce expression of the chemokine SDF-1, to alter the Ca2+ signaling, and to stimulate cell migration of glioblastoma cells. Here, we quantified fractionated IR-induced migration/brain infiltration of human glioblastoma cells in vitro and in an orthotopic mouse model and analyzed the role of SDF-1/CXCR4 signaling and BK channels. To this end, the radiation-induced migratory phenotypes of human T98G and far-red fluorescent U-87MG-Katushka glioblastoma cells were characterized by mRNA and protein expression, fura-2 Ca2+ imaging, BK patch-clamp recording and transfilter migration assay. In addition, U-87MG-Katushka cells were grown to solid glioblastomas in the right hemispheres of immunocompromised mice, fractionated irradiated (6 MV photons) with 5 × 0 or 5 × 2 Gy, and SDF-1, CXCR4, and BK protein expression by the tumor as well as glioblastoma brain infiltration was analyzed in dependence on BK channel targeting by systemic paxilline application concomitant to IR. As a result, IR stimulated SDF-1 signaling and induced migration of glioblastoma cells in vitro and in vivo. Importantly, paxilline blocked IR-induced migration in vivo. Collectively, our data demonstrate that fractionated IR of glioblastoma stimulates and BK K+ channel targeting mitigates migration and brain infiltration of glioblastoma cells in vivo. This suggests that BK channel targeting might represent a novel approach to overcome radiation-induced spreading of malignant brain tumors during radiotherapy.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Cell Movement/radiation effects , Glioblastoma/pathology , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , Apoptosis/radiation effects , Brain Neoplasms/metabolism , Brain Neoplasms/radiotherapy , Cell Proliferation/radiation effects , Chemokine CXCL12/metabolism , Female , Glioblastoma/metabolism , Glioblastoma/radiotherapy , Humans , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Radiation, Ionizing , Receptors, CXCR4/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
J Vis Exp ; (106): e53482, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26709831

ABSTRACT

The thymus, the primary organ for the generation of αß T cells and backbone of the adaptive immune system in vertebrates, has long been considered as the only source of αßT cells. Yet, thymic involution begins early in life leading to a drastically reduced output of naïve αßT cells into the periphery. Nevertheless, even centenarians can build immunity against newly acquired pathogens. Recent research suggests extrathymic αßT cell development, however our understanding of pathways that may compensate for thymic loss of function are still rudimental. γδ T cells are innate lymphocytes that constitute the main T-cell subset in the tissues. We recently ascribed a so far unappreciated outstanding function to a γδ T cell subset by showing that the scarce entity of CD4(+) Vδ1(+)γδ T cells can transdifferentiate into αßT cells in inflammatory conditions. Here, we provide the protocol for the isolation of this progenitor from peripheral blood and its subsequent cultivation. Vδ1 cells are positively enriched from PBMCs of healthy human donors using magnetic beads, followed by a second step wherein we target the scarce fraction of CD4(+) cells with a further magnetic labeling technique. The magnetic force of the second labeling exceeds the one of the first magnetic label, and thus allows the efficient, quantitative and specific positive isolation of the population of interest. We then introduce the technique and culture condition required for cloning and efficiently expanding the cells and for identification of the generated clones by FACS analysis. Thus, we provide a detailed protocol for the purification, culture and ex vivo expansion of CD4(+) Vδ1(+)γδ T cells. This knowledge is prerequisite for studies that relate to this αßT cell progenitor`s biology and for those who aim to identify the molecular triggers that are involved in its transdifferentiation.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Cell Culture Techniques/methods , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/physiology , Cell Transdifferentiation , Humans , Lymphocyte Activation , Stem Cells/cytology , Stem Cells/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology
19.
Ann Hematol ; 94(11): 1907-19, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26264693

ABSTRACT

GVHD, both acute and chronic, remains the major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation. Thus, there is still a great need for therapeutic tools for the prevention and treatment of GVHD. Several biologics have shown promising results in salvage therapies but are attendant on an increased risk for opportunistic infections, lymphoproliferative disorders, and relapse. This is partly due to efficient T cell elimination that neither dissects alloreactive from non-alloreactive T cells nor considers functional and structural distinctiveness of pathogen- and malignancy-reactive γδ and iNKT T cells. A novel, humanized monoclonal antibody, GZ-αßTCR, specific for the human αß T cell receptor, was evaluated in a xenogeneic GVHD model for its potential to prevent or ameliorate GVHD and prolong survival. We could show that GZ-αßTCR significantly attenuated clinical signs of GVHD and prolonged survival by preferential depletion of CD4 cells and the naïve T cell compartment, the trigger and driver of GVHD. In a regimen that included a preemptive dose, GZ-αßTCR treatment sufficiently abrogated GVHD. Importantly, GZ-αßTCR's specificity spared host cell-mediated immune competence of cell types other than αßT cells: namely γδT cells. GZ-αßTCR's outstanding capacity to prevent GVHD and ameliorate an ongoing GVHD while sparing immune cells other than αßT cells strongly recommends GZ-αßTCR for the prevention and treatment of acute GVHD in clinical settings.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Receptors, Antigen, T-Cell, alpha-beta/immunology , Transplantation Conditioning/methods , Vaccination/methods , Animals , Cells, Cultured , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Hematopoietic Stem Cell Transplantation/adverse effects , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, Transgenic , Survival Analysis
20.
Oncoimmunology ; 4(7): e1014760, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26140238

ABSTRACT

Stimulating the immune system to attack cancer is a promising approach, even for the control of advanced cancers. Several cytokines that promote interferon-γ-dominated immune responses show antitumor activity, with interleukin 12 (IL-12) being of major importance. Here, we used an antibody-IL-12 fusion protein (NHS-IL12) that binds histones of necrotic cells to treat human sarcoma in humanized mice. Following sarcoma engraftment, NHS-IL12 therapy was combined with either engineered IL-7 (FcIL-7) or IL-2 (IL-2MAB602) for continuous cytokine bioavailability. NHS-IL12 strongly induced innate and adaptive antitumor immunity when combined with IL-7 or IL-2. NHS-IL12 therapy significantly improved survival of sarcoma-bearing mice and caused long-term remissions when combined with IL-2. NHS-IL12 induced pronounced cancer cell senescence, as documented by strong expression of senescence-associated p16INK4a and nuclear translocation of p-HP1γ, and permanent arrest of cancer cell proliferation. In addition, this cancer immunotherapy initiated the induction of myogenic differentiation, further promoting the hypothesis that efficient antitumor immunity includes mechanisms different from cytotoxicity for efficient cancer control in vivo.

SELECTION OF CITATIONS
SEARCH DETAIL
...