Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Front Immunol ; 15: 1391954, 2024.
Article in English | MEDLINE | ID: mdl-38765008

ABSTRACT

Sarcomas are rare and heterogeneous malignancies that are difficult to treat. Approximately 50% of patients diagnosed with sarcoma develop metastatic disease with so far very limited treatment options. The transmembrane protein B7-H3 reportedly is expressed in various malignancies, including different sarcoma subtypes. In several cancer entities B7-H3 expression is associated with poor prognosis. In turn, B7-H3 is considered a promising target for immunotherapeutic approaches. We here report on the preclinical characterization of a B7-H3xCD3 bispecific antibody in an IgG-based format, termed CC-3, for treatment of different sarcoma subtypes. We found B7-H3 to be expressed on all sarcoma cells tested and expression on sarcoma patients correlated with decreased progression-free and overall survival. CC-3 was found to elicit robust T cell responses against multiple sarcoma subtypes, resulting in significant activation, release of cytokines and effector molecules. In addition, CC-3 promoted T cell proliferation and differentiation, resulting in the generation of memory T cell subsets. Finally, CC-3 induced potent target cell lysis in a target cell restricted manner. Based on these results, a clinical trial evaluating CC-3 in soft tissue sarcoma is currently in preparation.


Subject(s)
Antibodies, Bispecific , B7 Antigens , Sarcoma , Humans , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Sarcoma/immunology , Sarcoma/drug therapy , B7 Antigens/immunology , B7 Antigens/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Line, Tumor , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Female , Male , Animals , Lymphocyte Activation/immunology , Middle Aged , CD3 Complex/immunology , Aged , Cell Proliferation , Adult
2.
Blood Cancer J ; 14(1): 67, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38637557

ABSTRACT

Acute myeloid leukemia (AML) remains a therapeutic challenge despite recent therapeutic advances. Although monoclonal antibodies (mAbs) engaging natural killer (NK) cells via antibody-dependent cellular cytotoxicity (ADCC) hold promise in cancer therapy, almost none have received clinical approval for AML, so far. Recently, CD276 (B7-H3) has emerged as a promising target for AML immunotherapy, due to its high expression on leukemic blasts of AML patients. Here, we present the preclinical development of the Fc-optimized CD276 mAb 8H8_SDIE with enhanced CD16 affinity. We demonstrate that 8H8_SDIE specifically binds to CD276 on AML cell lines and primary AML cells and induces pronounced NK cell activation and degranulation as measured by CD69, CD25, and CD107a. Secretion of IFNγ, TNF, granzyme B, granulysin, and perforin, which mediate NK cell effector functions, was induced by 8H8_SDIE. A pronounced target cell-restricted lysis of AML cell lines and primary AML cells was observed in cytotoxicity assays using 8H8_SDIE. Finally, xenograft models with 8H8_SDIE did not cause off-target immune activation and effectively inhibited leukemia growth in vivo. We here present a novel attractive immunotherapeutic compound that potently induces anti-leukemic NK cell reactivity in vitro and in vivo as treatment option for AML.


Subject(s)
Killer Cells, Natural , Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/drug therapy , Antibody-Dependent Cell Cytotoxicity , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , B7 Antigens/metabolism , B7 Antigens/pharmacology
3.
Int J Mol Sci ; 25(7)2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38612935

ABSTRACT

Ligands of the natural killer group 2D (NKG2DL) family are expressed on malignant cells and are usually absent from healthy tissues. Recognition of NKG2DLs such as MICA/B and ULBP1-3 by the activating immunoreceptor NKG2D, expressed by NK and cytotoxic T cells, stimulates anti-tumor immunity in breast cancer. Upregulation of membrane-bound NKG2DLs in breast cancer has been demonstrated by immunohistochemistry. Tumor cells release NKG2DLs via proteolytic cleavage as soluble (s)NKG2DLs, which allows for effective immune escape and is associated with poor prognosis. In this study, we collected serum from 140 breast cancer (BC) and 20 ductal carcinoma in situ (DCIS) patients at the time of initial diagnosis and 20 healthy volunteers (HVs). Serum levels of sNKG2DLs were quantified through the use of ELISA and correlated with clinical data. The analyzed sNKG2DLs were low to absent in HVs and significantly higher in BC patients. For some of the ligands analyzed, higher sNKG2DLs serum levels were associated with the classification of malignant tumor (TNM) stage and grading. Low sMICA serum levels were associated with significantly longer progression-free (PFS) and overall survival (OS). In conclusion, we provide the first insights into sNKG2DLs in BC patients and suggest their potential role in tumor immune escape in breast cancer. Furthermore, our observations suggest that serum sMICA levels may serve as a prognostic parameter in the patients analyzed in this study.


Subject(s)
Breast Neoplasms , Carcinoma, Intraductal, Noninfiltrating , Humans , Female , Research Personnel , Enzyme-Linked Immunosorbent Assay , Health Status
4.
Sci Transl Med ; 16(737): eadh1988, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38446900

ABSTRACT

Despite the advances in cancer treatment achieved, for example, by the CD20 antibody rituximab, an urgent medical need remains to optimize the capacity of such antibodies to induce antibody-dependent cellular cytotoxicity (ADCC) that determines therapeutic efficacy. The cytokine IL-15 stimulates proliferation, activation, and cytolytic capacity of NK cells, but broad clinical use is prevented by short half-life, poor accumulation at the tumor site, and severe toxicity due to unspecific immune activation. We here report modified immunocytokines consisting of Fc-optimized CD19 and CD20 antibodies fused to an IL-15 moiety comprising an L45E-E46K double mutation (MIC+ format). The E46K mutation abrogated binding to IL-15Rα, thereby enabling substitution of physiological trans-presentation by target binding and thus conditional IL-15Rßγ stimulation, whereas the L45E mutation optimized IL-15Rßγ agonism and producibility. In vitro analysis of NK activation, anti-leukemia reactivity, and toxicity using autologous and allogeneic B cells confirmed target-dependent function of MIC+ constructs. Compared with Fc-optimized CD19 and CD20 antibodies, MIC+ constructs mediated superior target cell killing and NK cell proliferation. Mouse models using luciferase-expressing human NALM-6 lymphoma cells, patient acute lymphoblastic leukemia (ALL) cells, and murine EL-4 lymphoma cells transduced with human CD19/CD20 as targets and human and murine NK cells as effectors, respectively, confirmed superior and target-dependent anti-leukemic activity. In summary, MIC+ constructs combine the benefits of Fc-optimized antibodies and IL-15 cytokine activity and mediate superior NK cell immunity with potentially reduced side effects. They thus constitute a promising new immunotherapeutic approach shown here for B cell malignancies.


Subject(s)
Interleukin-15 , Lymphoma , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing , Antibodies , Antigens, CD19 , Cytokines , Immunoglobulin Fc Fragments
5.
Front Immunol ; 15: 1343929, 2024.
Article in English | MEDLINE | ID: mdl-38322253

ABSTRACT

Pancreatic cancer is a highly lethal disease with limited treatment options. Hence, there is a considerable medical need for novel treatment strategies. Monoclonal antibodies (mAbs) have significantly improved cancer therapy, primarily due to their ability to stimulate antibody-dependent cellular cytotoxicity (ADCC), which plays a crucial role in their therapeutic efficacy. As a result, significant effort has been focused on improving this critical function by engineering mAbs with Fc regions that have increased affinity for the Fc receptor CD16 expressed on natural killer (NK) cells, the major cell population that mediates ADCC in humans. Here we report on the preclinical characterization of a mAb directed to the target antigen B7-H3 (CD276) containing an Fc part with the amino acid substitutions S239D/I332E to increase affinity for CD16 (B7-H3-SDIE) for the treatment of pancreatic cancer. B7-H3 (CD276) is highly expressed in many tumor entities, whereas expression on healthy tissues is more limited. Our findings confirm high expression of B7-H3 on pancreatic cancer cells. Furthermore, our study shows that B7-H3-SDIE effectively activates NK cells against pancreatic cancer cells in an antigen-dependent manner, as demonstrated by the analysis of NK cell activation, degranulation and cytokine release. The activation of NK cells resulted in significant tumor cell lysis in both short-term and long-term cytotoxicity assays. In conclusion, B7-H3-SDIE constitutes a promising agent for the treatment of pancreatic cancer.


Subject(s)
Immunotherapy , Pancreatic Neoplasms , Humans , Immunotherapy/methods , Antibody-Dependent Cell Cytotoxicity , Antibodies, Monoclonal , Killer Cells, Natural , Pancreatic Neoplasms/metabolism , B7 Antigens/metabolism
6.
Int J Mol Sci ; 24(17)2023 Aug 24.
Article in English | MEDLINE | ID: mdl-37685962

ABSTRACT

Triple-negative breast cancer (TNBC) is a particularly aggressive subtype of breast cancer with a poor response rate to conventional systemic treatment and high relapse rates. Members of the natural killer group 2D ligand (NKG2DL) family are expressed on cancer cells but are typically absent from healthy tissues; thus, they are promising tumor antigens for novel immunotherapeutic approaches. We developed bispecific fusion proteins (BFPs) consisting of the NKG2D receptor domain targeting multiple NKG2DLs, fused to either anti-CD3 (NKG2D-CD3) or anti-CD16 (NKG2D-CD16) Fab fragments. First, we characterized the expression of the NKG2DLs (MICA, MICB, ULBP1-4) on TNBC cell lines and observed the highest surface expression for MICA and ULBP2. Targeting TNBC cells with NKG2D-CD3/CD16 efficiently activated both NK and T cells, leading to their degranulation and cytokine release and lysis of TNBC cells. Furthermore, PBMCs from TNBC patients currently undergoing chemotherapy showed significantly higher NK and T cell activation and tumor cell lysis when stimulated with NKG2D-CD3/CD16. In conclusions, BFPs activate and direct the NK and T cells of healthy and TNBC patients against TNBC cells, leading to efficient eradication of tumor cells. Therefore, NKG2D-based NK and T cell engagers could be a valuable addition to the treatment options for TNBC patients.


Subject(s)
Recombinant Fusion Proteins , Triple Negative Breast Neoplasms , Humans , Administration, Cutaneous , Aggression , Ligands , NK Cell Lectin-Like Receptor Subfamily K/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Recombinant Fusion Proteins/therapeutic use , Receptors, IgG , CD3 Complex
7.
Mol Ther ; 31(4): 1033-1045, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36793213

ABSTRACT

T cell-based immunotherapy has revolutionized oncological treatment. However, many patients do not respond to treatment, and long-term remissions remain rare, particularly in gastrointestinal cancers like colorectal cancer (CRC). B7-H3 is overexpressed in multiple cancer entities including CRC on both tumor cells and tumor vasculature, the latter facilitating influx of effector cells into the tumor site upon therapeutic targeting. We generated a panel of T cell-recruiting B7-H3xCD3 bispecific antibodies (bsAbs) and show that targeting a membrane-proximal B7-H3 epitope allows for a 100-fold reduction of CD3 affinity. In vitro, our lead compound CC-3 showed superior tumor cell killing, T cell activation, proliferation, and memory formation, whereas undesired cytokine release was reduced. In vivo, CC-3 mediated potent antitumor activity in three independent models using immunocompromised mice adoptively transferred with human effector cells with regard to prevention of lung metastasis and flank tumor growth as well as elimination of large established tumors. Thus, fine-tuning of both target and CD3 affinities as well as binding epitopes allowed for the generation of a B7-H3xCD3 bsAbs with promising therapeutic activity. CC-3 is presently undergoing good manufacturing practice (GMP) production to enable evaluation in a clinical "first-in-human" study in CRC.


Subject(s)
Antibodies, Bispecific , Gastrointestinal Neoplasms , Humans , Mice , Animals , Immunoglobulin G , T-Lymphocytes , Gastrointestinal Neoplasms/therapy , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Immunotherapy , Cell Line, Tumor
8.
Front Immunol ; 13: 1002898, 2022.
Article in English | MEDLINE | ID: mdl-36275693

ABSTRACT

Natural killer (NK) cells largely contribute to antibody-dependent cellular cytotoxicity (ADCC), a central factor for success of monoclonal antibodies (mAbs) treatment of cancer. The B7 family member B7-H3 (CD276) recently receives intense interest as a novel promising target antigen for immunotherapy. B7-H3 is highly expressed in many tumor entities, whereas expression on healthy tissues is rather limited. We here studied expression of B7-H3 in sarcoma, and found substantial levels to be expressed in various bone and soft-tissue sarcoma subtypes. To date, only few immunotherapeutic options for treatment of sarcomas that are limited to a minority of patients are available. We here used a B7-H3 mAb to generate chimeric mAbs containing either a wildtype Fc-part (8H8_WT) or a variant Fc part with amino-acid substitutions (S239D/I332E) to increase affinity for CD16 expressing NK cells (8H8_SDIE). In comparative studies we found that 8H8_SDIE triggers profound NK cell functions such as activation, degranulation, secretion of IFNγ and release of NK effector molecules, resulting in potent lysis of different sarcoma cells and primary sarcoma cells derived from patients. Our findings emphasize the potential of 8H8_SDIE as novel compound for treatment of sarcomas, particularly since B7-H3 is expressed in bone and soft-tissue sarcoma independent of their subtype.


Subject(s)
Killer Cells, Natural , Sarcoma , Humans , Immunoglobulin Fc Fragments , Antibody-Dependent Cell Cytotoxicity , Antibodies, Monoclonal , Sarcoma/therapy , Sarcoma/drug therapy , B7 Antigens/genetics
9.
Cancers (Basel) ; 14(16)2022 Aug 16.
Article in English | MEDLINE | ID: mdl-36010934

ABSTRACT

Antibodies against the B cell-specific antigens CD20 and CD19 have markedly improved the treatment of B cell-derived lymphoma and autoimmune diseases by depleting malignant and autoreactive B cells. However, since CD20 and CD19 are also expressed on healthy B cells, such antibodies lack disease specificity. Here, we optimize a previously developed concept that uses bispecific antibodies to induce apoptosis selectively in malignant and autoreactive B cells that express the death receptor CD95. We describe the development and characterization of bispecific antibodies with CD95xCD20 and CD95xCD19 specificity in a new IgG-based format. We could show that especially the CD95xCD20 antibody mediated a strong induction of apoptosis in malignant B cells in vitro. In vivo, the antibody was clearly superior to the previously used Fabsc format with identical specificities. In addition, both IgGsc antibodies depleted activated B cells in vitro, leading to a significant reduction in antibody production and cytokine secretion. The killing of resting B cells and hepatocytes that lack CD95 and CD20/CD19, respectively, was marginal. Thus, our results imply that bispecific anti-CD95 antibodies in the IgGsc format are an attractive tool for a more selective and efficient depletion of malignant as well as autoreactive B cells.

10.
Ann Hematol ; 101(4): 773-780, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35044512

ABSTRACT

Several genetic and clinical markers are established as prognostic factors in chronic lymphocytic leukemia (CLL). However, additional markers are needed for risk stratification. Flow cytometric analysis is a mainstay of CLL diagnostics, thus identification of novel prognostic surface markers can improve risk assessment without increasing burden for patients and physicians. Furthermore, surface molecules preferentially expressed in high-risk cases could serve as therapeutic targets for immunotherapy. CD105 (endoglin) is a TGF-beta coreceptor and activates endothelial cells in healthy tissues and cancer. In addition, it is expressed on healthy hematopoietic precursors as well as lymphoid and myeloid leukemias. In acute myeloid leukemia (AML), a CD105 antibody is successfully applied in clinical studies. In CLL, mRNA expression of the CD105 gene ENG reportedly correlates with other risk factors but failed to show significant correlation with overall survival. However, CD105 protein expression in CLL has never been studied. We here analyzed CD105 surface expression on CLL cells from 71 patients by flow cytometry and report for the first time that substantial levels of CD105 are detectable on CLL cells in 70.4% of patients. Using receiver operating characteristics, we established a cutoff of 5.99% positive cells to distinguish between low and high CD105 levels, the latter correlating with decreased time to first treatment and overall survival. High CD105 expression further correlates with CD38 expression. Our study identified membrane expression of CD105 as a potential risk marker and therapeutic target in high-risk CLL. However, multivariant analyses of large cohorts should be performed in confirmatory studies.


Subject(s)
Endoglin/analysis , Leukemia, Lymphocytic, Chronic, B-Cell , Leukemia, Myeloid, Acute , Endoglin/genetics , Endothelial Cells/metabolism , Flow Cytometry , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Myeloid, Acute/genetics , Prognosis
11.
Cancers (Basel) ; 13(18)2021 Sep 13.
Article in English | MEDLINE | ID: mdl-34572822

ABSTRACT

T cell-recruiting bispecific antibodies (bsAbs) are successfully used for the treatment of cancer. However, effective treatment with bsAbs is so far hampered by severe side effects, i.e., potentially life-threatening cytokine release syndrome. Off-target T cell activation due to binding of bispecific CD3 antibodies to T cells in the absence of target cells may contribute to excessive cytokine release. We report here, in an in vitro setting, that off-target T cell activation is induced by bsAbs with high CD3 binding affinity and increased by endothelial- or lymphoid cells that act as stimulating bystander cells. Blocking antibodies directed against the adhesion molecules CD18/CD54 or CD2/CD58 markedly reduced this type of off-target T cell activation. CD18 blockade-in contrast to CD2-did not affect the therapeutic activity of various bsAbs. Since CD18 antibodies have been shown to be safely applicable in patients, blockade of this integrin holds promise as a potential target for the prevention of unwanted off-target T cell activation and allows the application of truly effective bsAb doses.

12.
Front Immunol ; 12: 653081, 2021.
Article in English | MEDLINE | ID: mdl-33936075

ABSTRACT

Soft tissue sarcoma (STS) constitutes a rare group of heterogeneous malignancies. Effective treatment options for most subtypes of STS are still limited. As a result, especially in metastatic disease, prognosis is still dismal. The ligands for the activating immunoreceptor NKG2D (NKG2DL) are commonly expressed in STS, but generally absent in healthy tissues. This provides the rationale for utilization of NKG2DL as targets for immunotherapeutic approaches. We here report on the preclinical characterization of bispecific fusion proteins (BFP) consisting of the extracellular domain of the NKG2D receptor fused to Fab-fragments directed against CD3 (NKG2D-CD3) or CD16 (NKG2D-CD16) for treatment of STS. After characterization of NKG2DL expression patterns on various STS cell lines, we demonstrated that both NKG2D-CD16 and NKG2D-CD3 induce profound T and NK cell reactivity as revealed by analysis of activation, degranulation and secretion of IFNγ as well as granule associated proteins, resulting in potent target cell lysis. In addition, the stimulatory capacity of the constructs to induce T and NK cell activation was analyzed in heavily pretreated STS patients and found to be comparable to healthy donors. Our results emphasize the potential of NKG2D-CD3 and NKG2D-CD16 BFP to target STS even in an advanced disease.


Subject(s)
CD3 Complex/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily K/genetics , Receptors, IgG/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , Sarcoma/drug therapy , Adult , Aged , Antibodies, Bispecific/genetics , Antibodies, Bispecific/pharmacology , CD3 Complex/metabolism , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cell Line, Tumor , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Primary Cell Culture , Protein Domains/genetics , Receptors, IgG/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology , Sarcoma/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Young Adult
13.
Nat Immunol ; 22(1): 74-85, 2021 01.
Article in English | MEDLINE | ID: mdl-32999467

ABSTRACT

T cell immunity is central for the control of viral infections. To characterize T cell immunity, but also for the development of vaccines, identification of exact viral T cell epitopes is fundamental. Here we identify and characterize multiple dominant and subdominant SARS-CoV-2 HLA class I and HLA-DR peptides as potential T cell epitopes in COVID-19 convalescent and unexposed individuals. SARS-CoV-2-specific peptides enabled detection of post-infectious T cell immunity, even in seronegative convalescent individuals. Cross-reactive SARS-CoV-2 peptides revealed pre-existing T cell responses in 81% of unexposed individuals and validated similarity with common cold coronaviruses, providing a functional basis for heterologous immunity in SARS-CoV-2 infection. Diversity of SARS-CoV-2 T cell responses was associated with mild symptoms of COVID-19, providing evidence that immunity requires recognition of multiple epitopes. Together, the proposed SARS-CoV-2 T cell epitopes enable identification of heterologous and post-infectious T cell immunity and facilitate development of diagnostic, preventive and therapeutic measures for COVID-19.


Subject(s)
COVID-19/immunology , Epitopes, T-Lymphocyte/immunology , Peptides/immunology , SARS-CoV-2/immunology , T-Lymphocytes/immunology , Viral Vaccines/immunology , COVID-19/prevention & control , COVID-19/virology , Cross Reactions/immunology , HLA-DR Antigens/immunology , HLA-DR Antigens/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Immunologic Memory/immunology , SARS-CoV-2/physiology , T-Lymphocytes/metabolism , Viral Vaccines/administration & dosage
14.
EMBO Mol Med ; 13(2): e11902, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33372710

ABSTRACT

The prostate-specific membrane antigen (PSMA) has been demonstrated in numerous studies to be expressed specifically on prostate carcinoma cells and on the neovasculature of several other cancer entities. However, the simultaneous expression of PSMA on both, tumor cells as well as tumor vessels remains unclear, even if such "dual" expression would constitute an important asset to facilitate sufficient influx of effector cells to a given tumor site. We report here on the generation of a PSMA antibody, termed 10B3, which exerts superior dual reactivity on sections of prostate carcinoma and squamous cell carcinoma of the lung. 10B3 was used for the construction of T-cell recruiting bispecific PSMAxCD3 antibodies in Fab- and IgG-based formats, designated Fabsc and IgGsc, respectively. In vitro, both molecules exhibited comparable activity. In contrast, only the larger IgGsc molecule induced complete and durable elimination of established tumors in humanized mice due to favorable pharmacokinetic properties. Upon treatment of three patients with metastasized prostate carcinoma with the IgGsc reagent, marked activation of T cells and rapid reduction of elevated PSA levels were observed.


Subject(s)
Antibodies, Bispecific , Prostatic Neoplasms , Animals , Antigens, Surface , Humans , Immunoglobulin G , Male , Mice , Prostatic Neoplasms/drug therapy , T-Lymphocytes
15.
Leuk Lymphoma ; 61(10): 2466-2474, 2020 10.
Article in English | MEDLINE | ID: mdl-32543333

ABSTRACT

Risk assessment in acute myeloid leukemia (AML) mainly relies on (cyto-)genetic and morphologic features. Nonetheless, further markers are needed to allow for accurate risk stratification. Type I Fc gamma receptors (FcγRs) such as CD16, CD32, and CD64 play an important role in mediating immunomodulatory functions in different myeloid cell types as well as NK and B cells. We here evaluated expression of the three FcγR on peripheral blood AML blasts. Using flow cytometry, we found heterogeneous expression of the FcγR throughout the patient cohort. Correlation of expression levels with disease outcome revealed significantly shorter OS in patients with CD16+ blasts at first diagnosis. CD32 and CD64 expression showed no association with survival but correlated with a mature phenotype and FAB M6. Our data provide clear evidence for the value of immunophenotyping FcγR expression on leukemic cells using peripheral blood, which is rapidly available and improves risk stratification in AML.


Subject(s)
Leukemia, Myeloid, Acute , Receptors, IgG , Flow Cytometry , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Prognosis , Receptors, IgG/genetics
16.
Int J Hematol ; 112(1): 57-64, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32266669

ABSTRACT

Several genetic and molecular markers are general predictors of outcome in acute myeloid leukemia (AML), but only few are predictive of outcomes after allogeneic hematopoietic stem cell transplantation (HSCT). Novel markers are needed to improve treatment decisions regarding HSCT. CD105 (endoglin) is a type I transmembrane protein capable of activating endothelial cells. Moreover, CD105 mediates stem cell properties of hematopoietic stem cells and enables repopulation within the bone marrow. Expression of CD105 on vessels of solid tumors and on AML blasts is correlated with poor prognosis. We recently demonstrated that CD105 expression is an independent predictor of overall survival in AML. However, its role in patients receiving intensive treatment with consecutive allogenic transplantation has not been assessed. Using flow cytometry, we analyzed primary samples of 41 patients who underwent HSCT. Using the previously defined SFI cut-off of 5.2, we identified differences in CD105 expression regarding FAB classification and NCCN risk score. Moreover, we detected differences regarding transplant indication and WHO classification with regards to CD105 surface levels. In patients undergoing HSCT high CD105 expression correlated significantly with poor overall survival. We identify CD105 expression in AML as prognostic marker for outcome after HSCT in AML.


Subject(s)
Endoglin/genetics , Endoglin/metabolism , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Adult , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Female , Gene Expression , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Predictive Value of Tests , Prognosis , Risk , Risk Assessment , Survival Rate , Transplantation, Homologous , Treatment Outcome
17.
Ann Hematol ; 99(3): 477-486, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31965270

ABSTRACT

Genetic and morphological markers are well-established prognostic factors in acute myeloid leukemia (AML). However, further reliable markers are urgently needed to improve risk stratification in AML. CD318 (CDCP1) is a transmembrane protein which in solid tumors promotes formation of metastasis and correlates with poor survival. Despite its broad expression on hematological precursor cells, its prognostic significance in hematological malignancies so far remains unclear. Here, we evaluated the role of CD318 as novel prognostic marker in AML by immunophenotyping of leukemic blasts. Flow cytometric evaluation of CD318 on leukemic cells in 70 AML patients revealed a substantial expression in 40/70 (57%) of all cases. CD318 surface levels were significantly correlated with overall survival in patients receiving anthracycline-based induction therapy or best available alternative therapy. Using receiver-operating characteristics, we established a cut-off value to define CD318lo and CD318hi expression in both cohorts. Notably, high CD318 expression correlated inversely as prognostic marker in both treatment cohorts: as poor prognostic marker in patients receiving intense therapy, whereas upon palliative care it correlated with better outcome. In conclusion, FACS-based determination of CD318 expression may serve as novel prognostic factor depending on implemented therapy in AML patients.


Subject(s)
Antigens, Neoplasm/blood , Biomarkers, Tumor/blood , Blast Crisis , Cell Adhesion Molecules/blood , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute , Adult , Aged , Aged, 80 and over , Blast Crisis/blood , Blast Crisis/mortality , Blast Crisis/pathology , Disease-Free Survival , Female , Humans , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Survival Rate
18.
J Immunother Cancer ; 7(1): 143, 2019 05 29.
Article in English | MEDLINE | ID: mdl-31142382

ABSTRACT

BACKGROUND: Monoclonal antibodies (mAbs) mediate their effects in great part by inducing ADCC of NK cells, and multiple efforts aim to increase this function by engineering mAbs optimized Fc-parts. Even more potent antitumor immunity can be induced by strategies to stimulate T cells with their profoundly higher effector potential. However, upon increased immunostimulatory potential, the necessity to target highly tumor-specific antigens becomes critically important to reduce side effects. METHODS: We here report on bispecific fusion proteins (BFP) that target ligands of the immunoreceptor NKG2D (NKG2DL), which are widely expressed on malignant cells but generally absent on healthy tissue. They consist of the extracellular domain of NKG2D as targeting moiety fused to Fab-fragments of CD3 (NKG2D-CD3) or CD16 (NKG2D-CD16) antibodies. RESULTS: NKG2D-CD16 displayed increased affinity to the FcγRIII on NK cells compared to engineered Fc-parts, which are contained in optimized mAbs that presently undergo clinical evaluation. In line, NKG2D-CD16 induced superior activation, degranulation, IFN-γ production and lysis of acute myeloid leukemia (AML) cell lines and patient AML cells. NKG2D-CD3 in turn potently stimulated T cells, and comparison of efficacy over time revealed that NKG2D-CD16 was superior upon short term application, while NKG2D-CD3 mediated overall more potent effects which manifested after longer times. This can be attributed to treatment-induced proliferation of T cells but not NK cells. CONCLUSIONS: Taken together, we here introduce novel "antibody-like" BFP that take advantage of the highly tumor-restricted expression of NKG2DL and potently activate the reactivity of NK cells or T cells for immunotherapy of AML.


Subject(s)
Immunotherapy/methods , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , T-Lymphocytes/immunology , Humans , Leukemia, Myeloid, Acute/pathology , Membrane Fusion Proteins
19.
Neoplasia ; 20(11): 1150-1160, 2018 11.
Article in English | MEDLINE | ID: mdl-30300827

ABSTRACT

OX40 and its ligand are members of the TNF/TNF receptor superfamily, which includes various molecules influencing cellular signaling and function of both tumor and immune cells. The ability of OX40 to promote proliferation and differentiation of activated T cells fueled present attempts to modulate this immune checkpoint to reinforce antitumor immunity. While we recently found evidence for the involvement of OX40 in pathophysiology of acute myeloid leukemia including natural killer (NK) cell immunosurveillance, less is known on its role in acute lymphoblastic leukemia (ALL). In the present study, OX40 expression on ALL cells was significantly associated with positivity for the adverse risk factor BCR-ABL. In line, signaling via OX40 increased metabolic activity of primary ALL cells and resulted in release of cytokines involved in disease pathophysiology. Furthermore, interaction of ALL-expressed OX40 with its cognate ligand on NK cells stimulated ALL cell lysis. The data presented thus not only identify the yet unknown involvement of OX40/OX40L in ALL pathophysiology and NK cell immunosurveillance but also point to the necessity to thoroughly consider the consequences of modulating the OX40/OX40L molecule system beyond its effects on T cells when developing OX40-targeting approaches for cancer immunotherapy.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptors, OX40/genetics , Biomarkers, Tumor , Cell Line, Tumor , Cytokines/metabolism , Cytotoxicity, Immunologic , Energy Metabolism , Flow Cytometry , Fusion Proteins, bcr-abl/metabolism , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , OX40 Ligand/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prognosis , Protein Binding , Receptors, OX40/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
20.
Cancer Immunol Res ; 6(2): 209-221, 2018 02.
Article in English | MEDLINE | ID: mdl-29321210

ABSTRACT

The TNF receptor family member OX40 promotes activation and proliferation of T cells, which fuels efforts to modulate this immune checkpoint to reinforce antitumor immunity. Besides T cells, NK cells are a second cytotoxic lymphocyte subset that contributes to antitumor immunity, particularly in leukemia. Accordingly, these cells are being clinically evaluated for cancer treatment through multiple approaches, such as adoptive transfer of ex vivo expanded polyclonal NK cells (pNKC). Here, we analyzed whether and how OX40 and its ligand (OX40L) influence NK-cell function and antileukemia reactivity. We report that OX40 is expressed on leukemic blasts in a substantial percentage of patients with acute myeloid leukemia (AML) and that OX40 can, after stimulation with agonistic OX40 antibodies, mediate proliferation and release of cytokines that act as growth and survival factors for the leukemic cells. We also demonstrate that pNKC differentially express OX40L, depending on the protocol used for their generation. OX40L signaling promoted NK-cell activation, cytokine production, and cytotoxicity, and disruption of OX40-OX40L interaction impaired pNKC reactivity against primary AML cells. Together, our data implicate OX40/OX40L in disease pathophysiology of AML and in NK-cell immunosurveillance. Our findings indicate that effects of the OX40-OX40L receptor-ligand system in other immune cell subsets and also malignant cells should be taken into account when developing OX40-targeted approaches for cancer immunotherapy. Cancer Immunol Res; 6(2); 209-21. ©2018 AACR.


Subject(s)
Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/immunology , OX40 Ligand/immunology , Receptors, OX40/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cytokines/immunology , Humans , Immunologic Surveillance , Mice , Mice, Inbred C57BL , Receptors, OX40/agonists , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...