Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Hematol Oncol ; 14(1): 152, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34556152

ABSTRACT

Chimeric antigen receptor T-cell (CAR-T) therapy has shown tremendous success in eradicating hematologic malignancies. However, this success has not yet been extrapolated to solid tumors due to the limited infiltration and persistence of CAR-T cells in the tumor microenvironment (TME). In this study, we screened a novel anti-CD70 scFv and generated CD70 CAR-T cells that showed effective antitumor functions against CD70+ renal carcinoma cells (RCCs) both in vitro and in vivo. We further evaluated the effect and explored the molecular mechanism of a PARP inhibitor (PARPi) in CAR-T cell immunotherapy by administering the PARPi to mouse xenografts model derived from human RCC cells. Treatment with the PARPi promoted CAR-T cell infiltration by stimulating a chemokine milieu that promoted CAR-T cell recruitment and the modulation of immunosuppression in the TME. Moreover, our data demonstrate that PARPi modulates the TME by activating the cGAS-STING pathway, thereby altering the balance of immunostimulatory signaling and enabling low-dose CAR-T cell treatment to induce effective tumor regression. These data demonstrate the application of CD70 CAR-T cell therapeutic strategies for RCC and the cross-talk between targeting DNA damage responses and antitumor CAR-T cell therapy. These findings provide insight into the mechanisms of PARPis in CAR-T cell therapy for RCC and suggest a promising adjuvant therapeutic strategy for CAR-T cell therapy in solid tumors.


Subject(s)
CD27 Ligand/antagonists & inhibitors , Carcinoma, Renal Cell/therapy , Immunotherapy, Adoptive/methods , Kidney Neoplasms/therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Single-Chain Antibodies/therapeutic use , Animals , CD27 Ligand/immunology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/immunology , Cell Line, Tumor , DNA Damage/drug effects , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Membrane Proteins/immunology , Mice , Nucleotidyltransferases/immunology , Signal Transduction
2.
Cancer Sci ; 112(9): 3655-3668, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34117815

ABSTRACT

This study aimed to investigate the cytotoxicity of a cluster of differentiation 70 antibody-drug conjugate (CD70-ADC) against ovarian cancer in in vitro and in vivo xenograft models. CD70 expression was assessed in clinical samples by immunohistochemical analysis. Western blotting and fluorescence-activated cell sorting analyses were used to determine CD70 expression in the ovarian cancer cell lines A2780 and SKOV3, and in the cisplatin-resistant ovarian cancer cell lines A2780cisR and SKOV3cisR. CD70 expression after cisplatin exposure was determined in A2780 cells transfected with mock- or nuclear factor (NF)-κB-p65-small interfering RNA. We developed an ADC with an anti-CD70 monoclonal antibody linked to monomethyl auristatin F and investigated its cytotoxic effect. We examined 63 ovarian cancer clinical samples; 43 (68.3%) of them expressed CD70. Among patients with advanced stage disease (n = 50), those who received neoadjuvant chemotherapy were more likely to exhibit high CD70 expression compared to those who did not (55.6% [15/27] vs 17.4% [4/23], P < .01). CD70 expression was confirmed in A2780cisR, SKOV3, and SKOV3cisR cells. Notably, CD70 expression was induced after cisplatin treatment in A2780 mock cells but not in A2780-NF-κB-p65-silenced cells. CD70-ADC was cytotoxic to A2780cisR, SKOV3, and SKOV3cisR cells, with IC50 values ranging from 0.104 to 0.341 nmol/L. In A2780cisR and SKOV3cisR xenograft models, tumor growth in CD70-ADC treated mice was significantly inhibited compared to that in the control-ADC treated mice (A2780cisR: 32.0 vs 1639.0 mm3 , P < .01; SKOV3cisR: 232.2 vs 584.9 mm3 , P < .01). Platinum treatment induced CD70 expression in ovarian cancer cells. CD70-ADC may have potential therapeutic implications in the treatment of CD70 expressing ovarian cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , CD27 Ligand/metabolism , Cisplatin/administration & dosage , Immunoconjugates/administration & dosage , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Aged , Animals , CD27 Ligand/antagonists & inhibitors , CD27 Ligand/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Silencing , Humans , Mice , Middle Aged , Ovarian Neoplasms/pathology , Signal Transduction , Transcription Factor RelA/deficiency , Transcription Factor RelA/genetics , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
Exp Eye Res ; 199: 108190, 2020 10.
Article in English | MEDLINE | ID: mdl-32798537

ABSTRACT

PURPOSE: To determine whether the CD27/CD70 pathway plays a significant role in corneal allograft rejection by investigating the effect of blocking the CD27/CD70 pathway by anti-CD70 antibody on corneal allograft survival. METHODS: Orthotopic penetrating keratoplasty was performed using C57BL/6 donor grafts and BALB/c recipients. Expression of CD27 and CD70 on rejected cornea was examined by immunohistochemistry. Corneal transplant recipients received intraperitoneal injection of anti-CD70 antibody (FR70) or control rat IgG. Alloreactivity was measured by mixed lymphoid reaction (MLR) in recipients administered control rat IgG and those administered anti-CD70 antibody. Corneal expression of IFN-γ and IL-12 was also examined in both groups. Graft opacity was assessed over an 8-week period and graft survival was evaluated using Kaplan-Meier survival curves. Proportion of CD4+CD44+ memory T cells in lymph nodes was measured by flow cytometry. RESULTS: CD4+CD27+ cells and CD11c+CD70+ cells were present in rejected cornea. Anti-CD70 antibody administration suppressed alloreactivity in corneal allograft recipients, and inhibited IFN-γ expression in recipient cornea (p < 0.05). Anti-CD70 antibody suppressed opacity score of recipient cornea and prolonged corneal allograft survival (p < 0.05). Proportion of CD4+CD44+ memory T cells in recipient lymph nodes was reduced by anti-CD70 antibody treatment. CONCLUSION: The CD27/CD70 pathway plays a significant role in corneal allograft rejection by initiating alloreactive Th1 cells and preserving memory T cells. Anti-CD70 antibody administration prolongs corneal allograft survival indicating the potential therapeutic effect of CD27/CD70 pathway blockade on corneal allograft rejection.


Subject(s)
CD27 Ligand/antagonists & inhibitors , Cornea/metabolism , Corneal Transplantation , Graft Rejection/prevention & control , Graft Survival/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/antagonists & inhibitors , Allografts , Animals , CD27 Ligand/biosynthesis , Cornea/pathology , Disease Models, Animal , Graft Rejection/immunology , Graft Rejection/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis
4.
Theranostics ; 10(17): 7622-7634, 2020.
Article in English | MEDLINE | ID: mdl-32685008

ABSTRACT

Purpose: Given that heterogeneous expression and variants of antigens on solid tumors are responsible for relapse after chimeric antigen receptor (CAR)-T cell therapy, we hypothesized that combinatorial targeting two tumor-associated antigens would lessen this problem and enhance the antitumor activity of T cells. Methods: The co-expression level of CD70 and B7-H3 was analyzed in multiple tumor tissue samples. Further, two putative antigens were identified in The Cancer Genome Atlas and Gene Expression Profiling Interactive Analysis database. Two CD70 targeted CARs with different antigen binding domain, truncated CD27 and CD70 specific single-chain antibody fragment (scFv), were designed to screen a more suitable target-antigen binding moiety. Accordingly, we designed a bivalent tandem CAR (TanCAR) and further assessed the anti-tumor efficacy of TanCAR-T cells in vitro and in vivo. Results: Our results indicated that co-expression of CD70 and B7-H3 was observed on multiple tumor types including kidney, breast, esophageal, liver, colon cancer, glioma as well as melanoma. The CD70 targeted CAR-T cells with binding moiety of CD70 specific scFv exhibit a higher affinity and antitumor effect against CD70+ tumor cells. TanCAR-T cells induced enhanced ability of cytolysis and cytokine release over unispecific CAR-T cells when encountering tumor cells expressing two target-antigens. Further, low doses of TanCAR-T cells could also effectively control the lung cancer and melanoma xenografts and improved overall survival of the treated animals. Conclusion: TanCAR-T cells targeting CD70 and B7-H3 exhibit enhanced antitumor functionality and improve the problem of antigenic heterogeneity and variant in the treatment against solid tumor and melanoma.


Subject(s)
Immunotherapy, Adoptive/methods , Neoplasms/therapy , Single-Chain Antibodies/pharmacology , T-Lymphocytes/transplantation , Animals , Antigens, Neoplasm/immunology , B7 Antigens/antagonists & inhibitors , B7 Antigens/immunology , CD27 Ligand/antagonists & inhibitors , CD27 Ligand/immunology , Cell Line, Tumor , Female , Humans , Mice , Neoplasms/immunology , Receptors, Chimeric Antigen/immunology , Single-Chain Antibodies/therapeutic use , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
5.
Anticancer Res ; 40(8): 4471-4479, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32727777

ABSTRACT

BACKGROUND/AIM: Adult T-cell leukemia (ATL) is a hematological malignancy caused by infection with human T-cell leukemia virus type 1 (HTLV-1). Chemotherapy, antibody therapy, and bone marrow transplantation are used to treat this disease, however, median survival time has not been significantly improved. Our aim was to develop and evaluate a novel antibody-drug conjugate (ADC) with regards to cell cytotoxicity and target specificity. MATERIALS AND METHODS: In this study, we have constructed a novel ADC, which is composed of an anti-CD70 single chain Fv-Fc antibody conjugated with the anticancer agent emtansine using a novel antibody modification method. Cell cytotoxicity and target specificity were assessed using a cell proliferation assay. RESULTS: The anti-CD70 ADC selectively killed HTLV-1-infected cells and ATL cells without affecting other cells. CONCLUSION: The anti-CD70 ADC offers some chemotherapeutic potential for the treatment of ATL.


Subject(s)
CD27 Ligand/antagonists & inhibitors , Immunoconjugates/pharmacology , Leukemia-Lymphoma, Adult T-Cell/immunology , Maytansine/pharmacology , Single-Chain Antibodies/pharmacology , Adult , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Jurkat Cells , Leukemia-Lymphoma, Adult T-Cell/therapy , Male , Middle Aged
6.
Nat Med ; 26(9): 1459-1467, 2020 09.
Article in English | MEDLINE | ID: mdl-32601337

ABSTRACT

Acute myeloid leukemia (AML) is driven by leukemia stem cells (LSCs) that resist conventional chemotherapy and are the major cause of relapse1,2. Hypomethylating agents (HMAs) are the standard of care in the treatment of older or unfit patients with AML, but responses are modest and not durable3-5. Here we demonstrate that LSCs upregulate the tumor necrosis factor family ligand CD70 in response to HMA treatment resulting in increased CD70/CD27 signaling. Blocking CD70/CD27 signaling and targeting CD70-expressing LSCs with cusatuzumab, a human αCD70 monoclonal antibody with enhanced antibody-dependent cellular cytotoxicity activity, eliminated LSCs in vitro and in xenotransplantation experiments. Based on these preclinical results, we performed a phase 1/2 trial in previously untreated older patients with AML with a single dose of cusatuzumab monotherapy followed by a combination therapy with the HMA azacitidine ( NCT03030612 ). We report results from the phase 1 dose escalation part of the clinical trial. Hematological responses in the 12 patients enrolled included 8 complete remission, 2 complete remission with incomplete blood count recovery and 2 partial remission with 4 patients achieving minimal residual disease negativity by flow cytometry at <10-3. Median time to response was 3.3 months. Median progression-free survival was not reached yet at the time of the data cutoff. No dose-limiting toxicities were reported and the maximum tolerated dose of cusatuzumab was not reached. Importantly, cusatuzumab treatment substantially reduced LSCs and triggered gene signatures related to myeloid differentiation and apoptosis.


Subject(s)
Antineoplastic Agents/therapeutic use , CD27 Ligand/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Neoplastic Stem Cells/drug effects , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Azacitidine/therapeutic use , DNA Methylation/drug effects , DNA Methylation/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Treatment Outcome , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
7.
Aging (Albany NY) ; 12(12): 12032-12050, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32559178

ABSTRACT

Aging is associated with immune dysregulation, especially T cell disorders, which result in increased susceptibility to various diseases. Previous studies have shown that loss of co-stimulatory receptors or accumulation of co-inhibitory molecules play important roles in T cell aging. In the present study, CD70, which was generally regarded as a costimulatory molecule, was found to be upregulated on CD4+ and CD8+ T cells of elderly individuals. Aged CD70+ T cells displayed a phenotype of over-activation, and expressed enhanced levels of numerous inhibitory receptors including PD-1, 2B4 and LAG-3. CD70+ T cells from elderly individuals exhibited increased susceptibility to apoptosis and high levels of inflammatory cytokines. Importantly, the functional dysregulation of CD70+ T cells associated with aging was reversed by blocking CD70. Collectively, this study demonstrated CD70 as a prominent regulator involved in immunosenescence, which led to defects and overwhelming inflammatory responses of T cells during aging. These findings provide a strong rationale for targeting CD70 to prevent dysregulation related to immunosenescence.


Subject(s)
CD27 Ligand/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunosenescence/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Apoptosis/drug effects , Apoptosis/immunology , CD27 Ligand/antagonists & inhibitors , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Female , Flow Cytometry , Healthy Volunteers , Humans , Immunosenescence/drug effects , Lymphocyte Activation/drug effects , Male , Middle Aged , Primary Cell Culture , Up-Regulation/immunology , Young Adult
8.
Front Immunol ; 11: 555996, 2020.
Article in English | MEDLINE | ID: mdl-33737923

ABSTRACT

Allograft rejection has been an obstacle for the long-term survival of patients. CD70, a tumor necrosis factor (TNF) family member critically expressed on antigen-presenting cells and strongly but transiently up-regulated during lymphocyte activation, represents an important co-stimulatory molecule that induces effective T cell responses. We used a mouse heterotopic cardiac transplantation model to evaluate the effects of monotherapy with the antibody targeting mouse CD70 (FR70) on transplantation tolerance and its immunoregulatory activity. FR70-treated C3H recipient mice permanently accepted B6 fully mismatched cardiac allografts. Consistent with the graft survival, the infiltration of CD8+ T cells in the graft was reduced, dendritic cells were differentiated into a tolerogenic status, and the number of regulatory T cells was elevated both in the graft and the recipient's spleen. In addition, naïve C3H given an adoptive transfer of spleen cells from the primary recipients with FR70 treatment accepted a heart graft from a matching B6 donor but not third-party BALB/c mice. Our findings show that treatment with FR70 induced regulatory cells and inhibited cytotoxic T cell proliferation, which led to long-term acceptance of mouse cardiac allografts. These findings highlight the potential role of anti-CD70 antibodies as a clinically effective treatment for allograft rejection.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD27 Ligand/antagonists & inhibitors , Dendritic Cells/immunology , Dendritic Cells/metabolism , Heart Transplantation , Transplantation Tolerance/drug effects , Transplantation Tolerance/immunology , Adoptive Transfer , Allografts , Animals , Antibodies, Monoclonal/pharmacology , Graft Rejection/immunology , Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Heart Transplantation/methods , Immunohistochemistry , Immunomodulation , Immunophenotyping , Mice , Spleen/immunology , Spleen/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
9.
Invest New Drugs ; 37(2): 297-306, 2019 04.
Article in English | MEDLINE | ID: mdl-30132271

ABSTRACT

Purpose This first-in-human study evaluated SGN-CD70A, an antibody-drug conjugate (ADC) directed against the integral plasma membrane protein CD70 and linked to a pyrrolobenzodiazepine (PBD) dimer, in patients with relapsed or refractory (R/R) CD70-positive non-Hodgkin lymphoma (NHL) including diffuse large B cell lymphoma (DLBCL), mantle cell lymphoma (MCL), and Grade 3b follicular lymphoma (FL3b). Methods SGN-CD70A was administered intravenously on Day 1 of 3-week cycles beginning at 8 mcg/kg with planned dose escalation to 200 mcg/kg. Due to observations of prolonged thrombocytopenia, the study was amended to dose every 6 weeks (q6wk). Results Twenty patients were enrolled and treated with SGN-CD70A. The maximum tolerated dose of SGN-CD70A was 30 mcg/kg q6wk. The most common adverse events (AEs) reported were thrombocytopenia (75%), nausea (55%), anemia (50%), and fatigue (50%). The onset for treatment-related thrombocytopenia typically occurred during Cycle 1. Most of the treatment-related events of thrombocytopenia were ≥ Grade 3. Antitumor activity in patients included 1 complete remission (CR) and 3 partial remissions (PRs), 2 of which were ongoing for at least 42.9 weeks. SGN-CD70A exposures were approximately dose proportional, with a mean terminal half-life of 3 to 5 days. Conclusions While modest single-agent activity was observed in heavily pretreated NHL patients, the applicability of SGN-CD70A is limited by the frequency and severity of thrombocytopenia, despite the long-term response with limited drug exposure.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD27 Ligand/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Lymphoma, B-Cell/drug therapy , Lymphoma, Mantle-Cell/drug therapy , Neoplasm Recurrence, Local/drug therapy , Salvage Therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/pharmacokinetics , Benzodiazepines/chemistry , Biomarkers, Tumor/metabolism , CD27 Ligand/immunology , CD27 Ligand/metabolism , Drug Resistance, Neoplasm/drug effects , Female , Follow-Up Studies , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Mantle-Cell/immunology , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/pathology , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Prognosis , Pyrroles/chemistry , Tissue Distribution
10.
Clin Cancer Res ; 23(21): 6411-6420, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28765328

ABSTRACT

Purpose: The purpose of this study was to evaluate safety, pharmacokinetics, pharmacodynamics, and preliminary antitumor efficacy of ARGX-110, a glyco-engineered monoclonal antibody, targeting CD70, in patients with CD70 expressing advanced malignancies.Experimental Design: Dose escalation with a sequential 3+3 design was performed in five steps at the 0.1, 1, 2, 5, and 10 mg/kg dose levels (N = 26). ARGX-110 was administered intravenously every 3 weeks until progression or intolerable toxicity. Dose-limiting toxicity was evaluated in the 21 days following the first ARGX-110 administration (Cycle 1). Samples for pharmacokinetics and pharmacodynamics were collected.Results: Dose-limiting toxicity was not observed and the maximum tolerated dose was not reached. ARGX-110 was generally well tolerated, with no dose-related increase in treatment-emergent adverse events (TEAE). The most common TEAE were fatigue and drug related infusion-related reactions (IRR). Of the 20 SAEs reported, five events, all IRRs, were considered related to ARGX-110. ARGX-110 demonstrates dose proportionality over the dose range 1 to 10 mg/kg, but not at 0.1 mg/kg and a terminal half-life of 10 to 13 days. The best overall response was stable disease (14/26) in all 26 evaluable patients with various malignancies and the mean duration of treatment was 15 weeks. No dose-response related antitumor activity was observed, but biomarker readouts provided signs of biological activity, particularly in patients with hematologic malignancies.Conclusions: This dose-escalation phase I trial provides evidence of good tolerability of ARGX-110, pharmacokinetics, and preliminary antitumor activity at all dose levels in generally heavily pretreated patients with advanced CD70-positive malignancies. Clin Cancer Res; 23(21); 6411-20. ©2017 AACR.


Subject(s)
Antibodies, Anti-Idiotypic/administration & dosage , Antibodies, Monoclonal/administration & dosage , CD27 Ligand/immunology , Neoplasms/drug therapy , Adult , Aged , Antibodies, Anti-Idiotypic/adverse effects , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , CD27 Ligand/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug-Related Side Effects and Adverse Reactions/classification , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/immunology , Neoplasms/pathology
11.
Nat Rev Rheumatol ; 13(4): 217-233, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28275260

ABSTRACT

TNF blockers are highly efficacious at dampening inflammation and reducing symptoms in rheumatic diseases such as rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis, and also in nonrheumatic syndromes such as inflammatory bowel disease. As TNF belongs to a superfamily of 19 structurally related proteins that have both proinflammatory and anti-inflammatory activity, reagents that disrupt the interaction between proinflammatory TNF family cytokines and their receptors, or agonize the anti-inflammatory receptors, are being considered for the treatment of rheumatic diseases. Biologic agents that block B cell activating factor (BAFF) and receptor activator of nuclear factor-κB ligand (RANKL) have been approved for the treatment of systemic lupus erythematosus and osteoporosis, respectively. In this Review, we focus on additional members of the TNF superfamily that could be relevant for the pathogenesis of rheumatic disease, including those that can strongly promote activity of immune cells or increase activity of tissue cells, as well as those that promote death pathways and might limit inflammation. We examine preclinical mouse and human data linking these molecules to the control of damage in the joints, muscle, bone or other tissues, and discuss their potential as targets for future therapy of rheumatic diseases.


Subject(s)
Molecular Targeted Therapy , Rheumatic Diseases/drug therapy , Rheumatic Diseases/immunology , Tumor Necrosis Factor Inhibitors , Tumor Necrosis Factors/metabolism , 4-1BB Ligand/antagonists & inhibitors , 4-1BB Ligand/metabolism , Animals , CD27 Ligand/antagonists & inhibitors , CD27 Ligand/metabolism , CD40 Ligand/antagonists & inhibitors , CD40 Ligand/metabolism , Cell Death , Cytokine TWEAK , Dendritic Cells/immunology , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/metabolism , Humans , Immune Tolerance , Lymphocyte Activation , Lymphotoxin-alpha/antagonists & inhibitors , Lymphotoxin-alpha/metabolism , OX40 Ligand/antagonists & inhibitors , OX40 Ligand/metabolism , Signal Transduction , T-Lymphocytes/immunology , TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/antagonists & inhibitors , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/antagonists & inhibitors , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Tumor Necrosis Factors/immunology
12.
J Exp Med ; 214(2): 359-380, 2017 02.
Article in English | MEDLINE | ID: mdl-28031480

ABSTRACT

Aberrant proliferation, symmetric self-renewal, increased survival, and defective differentiation of malignant blasts are key oncogenic drivers in acute myeloid leukemia (AML). Stem cell gene signatures predict poor prognosis in AML patients; however, with few exceptions, these deregulated molecular pathways cannot be targeted therapeutically. In this study, we demonstrate that the TNF superfamily ligand-receptor pair CD70/CD27 is expressed on AML blasts and AML stem/progenitor cells. CD70/CD27 signaling in AML cells activates stem cell gene expression programs, including the Wnt pathway, and promotes symmetric cell divisions and proliferation. Soluble CD27, reflecting the extent of CD70/CD27 interactions in vivo, was significantly elevated in the sera of newly diagnosed AML patients and is a strong independent negative prognostic biomarker for overall survival. Blocking the CD70/CD27 interaction by mAb induced asymmetric cell divisions and differentiation in AML blasts and AML stem/progenitor cells, inhibited cell growth and colony formation, and significantly prolonged survival in murine AML xenografts. Importantly, hematopoietic stem/progenitor cells from healthy BM donors express neither CD70 nor CD27 and were unaffected by blocking mAb treatment. Therefore, targeting CD70/CD27 signaling represents a promising therapeutic strategy for AML.


Subject(s)
Blast Crisis/etiology , CD27 Ligand/physiology , Leukemia, Myeloid, Acute/pathology , Signal Transduction/physiology , Tumor Necrosis Factor Receptor Superfamily, Member 7/physiology , Aged , Animals , Antibodies, Monoclonal/therapeutic use , CD27 Ligand/antagonists & inhibitors , Germinal Center Kinases , Humans , Leukemia, Myeloid, Acute/drug therapy , Mice , Middle Aged , Protein Serine-Threonine Kinases/physiology , TNF Receptor-Associated Factor 2/physiology , Tumor Cells, Cultured , Tumor Necrosis Factor Receptor Superfamily, Member 7/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 7/blood , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Wnt Signaling Pathway/physiology
13.
Biopharm Drug Dispos ; 37(2): 93-106, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25869904

ABSTRACT

CD70 is a tumor necrosis factor (TNF)-like type II integral membrane protein that is transiently expressed on activated T- and B-lymphocytes. Aberrant expression of CD70 was identified in both solid tumors and haematologic malignancies. BMS-936561 (αCD70_MED-A) is an antibody-drug conjugate composed of a fully human anti-CD70 monoclonal antibody (αCD70) conjugated with a duocarmycin derivative, MED-A, through a maleimide-containing citrulline-valine dipeptide linker. MED-A is a carbamate prodrug that is activated by carboxylesterase to its active form, MED-B, to exert its DNA alkylation activity. In vitro serum stability studies suggested the efficiencies of hydrolyzing the carbamate-protecting group in αCD70_MED-A followed a rank order of mouse>rat > >monkey>dog~human. Pharmacokinetics of αCD70_MED-A was evaluated in mice, monkeys, and dogs after single intravenous doses. In mice, αCD70_MED-A was cleared rapidly, with no detectable exposures after 15 min following dosing. In contrast, αCD70_MED-A was much more stable in monkeys and dogs. The clearance of αCD70_MED-A in monkeys was 58 mL/d/kg, ~2-fold faster than that in dogs (31 mL/d/kg). The human PK profiles of the total αCD70 and αCD70_MED-A were predicted using allometrically scaled monkeys PK parameters of αCD70 and the carbamate hydrolysis rate constant estimated in dogs. Comparing the predicted and observed human PK from the phase I study, the dose-normalized concentration-time profiles of αCD70_MED-A and the total αCD70 were largely within the 5(th)-95(th) percentile of the predicted profiles.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents, Alkylating/pharmacokinetics , CD27 Ligand/antagonists & inhibitors , Immunoconjugates/pharmacokinetics , Indoles/pharmacokinetics , Prodrugs/pharmacokinetics , Animals , Antibodies, Monoclonal/blood , Antineoplastic Agents, Alkylating/blood , CD27 Ligand/immunology , Dogs , Humans , Immunoconjugates/blood , Indoles/blood , Macaca fascicularis , Mice, Inbred BALB C , Models, Biological
14.
Immunotherapy ; 7(6): 655-67, 2015.
Article in English | MEDLINE | ID: mdl-26098609

ABSTRACT

In 2013, cancer immunotherapy was named 'breakthrough of the year' based on the outcome of clinical trials with blocking antibodies to the T-cell co-inhibitory receptors CTLA-4 and PD-1. This success has emphasized that cytotoxic T-cell responses to cancer can occur, but are limited by peripheral tolerance and by immunosuppression in the tumor microenvironment. Targeting of CTLA-4, PD-1 or its ligands partly overcomes these limitations and can now be applied in multiple immunogenic cancer types. Furthermore, an increased success rate is expected from combining CTLA-4 and/or PD-1 blocking with deliberate engagement of T-cell co-stimulatory receptors, particularly TNF receptor (R) family members. The TNFR family includes CD27 (Tnfrsf7), for which an agonistic antibody has recently entered clinical trials. In this review, we describe how CD27 co-stimulation impacts the T-cell response, with the purpose to illuminate how CD27 agonism can be exploited in cancer immunotherapy.


Subject(s)
Antibodies, Neutralizing/therapeutic use , CD27 Ligand/antagonists & inhibitors , Immunotherapy/methods , Neoplasms/therapy , Signal Transduction/drug effects , Tumor Necrosis Factor Receptor Superfamily, Member 7/antagonists & inhibitors , Animals , CD27 Ligand/immunology , CTLA-4 Antigen/immunology , Humans , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/immunology , Signal Transduction/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
15.
PLoS One ; 8(4): e61446, 2013.
Article in English | MEDLINE | ID: mdl-23613854

ABSTRACT

Metastatic renal cell carcinoma (RCC) is an incurable disease in clear need of new therapeutic interventions. In early-phase clinical trials, the cytokine IFN-γ showed promise as a biotherapeutic for advanced RCC, but subsequent trials were less promising. These trials, however, focused on the indirect immunomodulatory properties of IFN-γ, and its direct anti-tumor effects, including its ability to kill tumor cells, remains mostly unexploited. We have previously shown that IFN-γ induces RIP1 kinase-dependent necrosis in cells lacking NF-κB survival signaling. RCC cells display basally-elevated NF-κB activity, and inhibiting NF-κB in these cells, for example by using the small-molecule proteasome blocker bortezomib, sensitizes them to RIP1-dependent necrotic death following exposure to IFN-γ. While these observations suggest that IFN-γ-mediated direct tumoricidal activity will have therapeutic benefit in RCC, they cannot be effectively exploited unless IFN-γ is targeted to tumor cells in vivo. Here, we describe the generation and characterization of two novel 'immunocytokine' chimeric proteins, in which either human or murine IFN-γ is fused to an antibody targeting the putative metastatic RCC biomarker CD70. These immunocytokines display high levels of species-specific IFN-γ activity and selective binding to CD70 on human RCC cells. Importantly, the IFN-γ immunocytokines function as well as native IFN-γ in inducing RIP1-dependent necrosis in RCC cells, when deployed in the presence of bortezomib. These results provide a foundation for the in vivo exploitation of IFN-γ-driven tumoricidal activity in RCC.


Subject(s)
CD27 Ligand/antagonists & inhibitors , Carcinoma, Renal Cell/pathology , Interferon-gamma/pharmacology , Kidney Neoplasms/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Antiviral Agents/pharmacology , Boronic Acids/pharmacology , Bortezomib , CD27 Ligand/metabolism , Carcinoma, Renal Cell/enzymology , Cell Death/drug effects , Cell Line, Tumor , Humans , Kidney Neoplasms/enzymology , Mice , Necrosis , Phosphorylation/drug effects , Protein Binding/drug effects , Pyrazines/pharmacology , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Species Specificity
17.
Immunol Lett ; 138(2): 122-8, 2011 Aug 30.
Article in English | MEDLINE | ID: mdl-21477620

ABSTRACT

Donor-reactive memory T cells mediated accelerated rejection is known as a barrier to the survival of transplanted organs. We investigated the combination of different monoclonal antibodies (mAbs) and donor-specific transfusion (DST) in memory T cells-based adoptive mice model. In the presence of donor-reactive memory T cells, the mean survival time (MST) of grafts in the anti-CD40L/LFA-1/DST group was 49.8d. Adding anti-CD44/CD70 mAbs to anti-CD40L/LFA-1/DST treatment. The MST was more than 100 d (MST>100 d). Compared with anti-CD40L/LFA-1/DST group, anti-CD40L/LFA-1/CD44/CD70/DST group notably reduced the expansion of memory T cells, enhanced the proportion of CD4+Foxp3+ regulatory T cells (Tregs) and suppressed donor-specific responses. Our data suggest that anti-CD40L/LFA-1/CD44/CD70mAbs and DST can synergistically inhibit accelerated rejection mediated by memory T cells to induce long-lived heart allograft acceptance in mice.


Subject(s)
Antibodies, Monoclonal , Graft Rejection , Graft Survival/immunology , Heart Transplantation , Immunomodulation , Isoantigens/immunology , Monocytes , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD27 Ligand/antagonists & inhibitors , CD27 Ligand/immunology , CD40 Ligand/antagonists & inhibitors , CD40 Ligand/immunology , Cell Proliferation/drug effects , Female , Graft Rejection/immunology , Graft Rejection/prevention & control , Heart Transplantation/immunology , Hyaluronan Receptors/immunology , Immunologic Memory , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/transplantation , Skin Transplantation/immunology , Spleen/cytology , Spleen/immunology , Transplantation, Homologous
18.
J Immunol ; 185(12): 7274-84, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21078900

ABSTRACT

CD70 is expressed in normal activated immune cells as well as in several types of tumors. It has been established that anti-CD70 mAb induces complement-dependent death of CD70(+) tumor cells, but how anti-CD70 mAb affects the intrinsic signaling is poorly defined. In this report, we show that ligation of CD70 expressed on EBV-transformed B cells using anti-CD70 mAb induced production of reactive oxygen species (ROS) and subsequent apoptosis. We observed an early expression of endoplasmic reticulum (ER) stress response genes that preceded the release of apoptotic molecules from the mitochondria and the cleavage of caspases. CD70-induced apoptosis was inhibited by pretreatment with the ER stress inhibitor salubrinal, ROS quencher N-acetylcysteine, and Ca(2+) chelator BAPTA. We supposed that ROS generation might be the first event of CD70-induced apoptosis because N-acetylcysteine blocked increases of ROS and Ca(2+), but BAPTA did not block ROS generation. We also found that CD70 stimulation activated JNK and p38 MAPK. JNK inhibitor SP600125 and p38 inhibitor SB203580 effectively blocked upregulation of ER stress-related genes and cleavage of caspases. Inhibition of ROS generation completely blocked phosphorylation of JNK and p38 MAPK and induction of ER stress-related genes. Taken together, we concluded that cross-linking of CD70 on EBV-transformed B cells triggered ER stress-mediated apoptosis via ROS generation and JNK and p38 MAPK pathway activation. Our report reveals alternate mechanisms of direct apoptosis through CD70 signaling and provides data supporting CD70 as a viable target for an Ab-based therapy against EBV-related tumors.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/immunology , CD27 Ligand/immunology , Endoplasmic Reticulum/immunology , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , MAP Kinase Kinase 4/immunology , Reactive Oxygen Species/immunology , Signal Transduction/immunology , Unfolded Protein Response/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Acetylcysteine/pharmacology , Animals , Anthracenes/pharmacology , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/virology , CD27 Ligand/antagonists & inhibitors , CD27 Ligand/metabolism , Callithrix , Cell Line, Transformed , Chelating Agents , Cinnamates/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Endoplasmic Reticulum/metabolism , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/metabolism , Free Radical Scavengers/pharmacology , Herpesvirus 4, Human/metabolism , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/metabolism , Phosphorylation/drug effects , Phosphorylation/immunology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Thiourea/analogs & derivatives , Thiourea/pharmacology , Unfolded Protein Response/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
19.
J Clin Periodontol ; 37(5): 412-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20507366

ABSTRACT

AIM: We sought to determine whether triclosan (2,4,4'-trichloro-2'-hydroxydiphenylether), an extensively used anti-plaque agent with broad-spectrum anti-microbial activity, with reported anti-inflammatory effects via inhibition of prostaglandin E2 and interleukin 1 (IL-1)beta, could also more broadly suppress multiple inflammatory gene pathways responsible for the pathogenesis of gingivitis and periodontitis. MATERIALS AND METHODS: As an exploratory study, the effects of triclosan on the inflammatory gene expression profile were assessed ex vivo using peripheral whole blood samples from eight periodontally healthy donors. Ten-millilitres whole blood aliquots were incubated 2 h with 0.3 microg/ml Escherichia coli lipopolysaccharide (LPS) with or without 0.5 microg/ml triclosan. Affymetrix microarray gene expression profiles from isolated leucocytes and pathway-specific quantitative polymerase chain reaction arrays were used to investigate changes in expression of target cytokines and cell signalling molecules. RESULTS: Ex vivo human whole blood assays indicated that triclosan significantly down-regulated the LPS-stimulated expression of Toll-like receptor signalling molecules and other multiple inflammatory molecules including IL-1 and IL-6 and the dampening of signals that activate the T-helper type 1 acquired immune response via suppression of CD70 with concomitant up-regulation of growth factors related to bone morphogenetic protein (BMP)2 and BMP6 synthesis. CONCLUSIONS: Anti-inflammatory effects were found in this exploratory survey, including suppression of microbial-pathogen recognition pathway molecules and the suppression of acute and chronic mediators of inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Gene Expression Profiling , Inflammation Mediators/antagonists & inhibitors , Inflammation/genetics , Toll-Like Receptors/genetics , Triclosan/pharmacology , Acute Disease , Adult , Bone Morphogenetic Protein 2/biosynthesis , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 6/biosynthesis , Bone Morphogenetic Protein 6/genetics , CD27 Ligand/antagonists & inhibitors , Chronic Disease , Female , Host-Pathogen Interactions/drug effects , Humans , Interleukin-1/antagonists & inhibitors , Interleukin-1/biosynthesis , Interleukin-1/genetics , Interleukin-6/antagonists & inhibitors , Interleukin-6/biosynthesis , Interleukin-6/genetics , Lipopolysaccharides/pharmacology , Male , Oligonucleotide Array Sequence Analysis , Signal Transduction/drug effects , Th1 Cells/immunology , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/biosynthesis , Young Adult
20.
Adv Exp Med Biol ; 647: 108-19, 2009.
Article in English | MEDLINE | ID: mdl-19760069

ABSTRACT

Expression of CD70, a member of the tumor necrosis factor superfamily, is restricted to activated T-and B-lymphocytes and mature dendritic cells. Binding of CD70 to its receptor, CD27, is important in priming, effector functions, differentiation and memory formation of T-cells as well as plasma and memory B-cell generation. Antibody blockade of CD70-CD27 interaction inhibits the onset of experimental autoimmune encephalomyelits and cardiac allograft rejection in mice. CD70 has been also detected on hematological tumors and on carcinomas. The highly restricted expression pattern of CD70 in normal tissues and its widespread expression in various malignancies as well as its potential role in autoimmune and inflammatory conditions makes it an attractive target for antibody-based therapeutics. This chapter provides an overview of the physiological role of CD70-CD27 interactions and discusses various approaches to target this pathway for therapeutic use in cancers and autoimmunity.


Subject(s)
Autoimmune Diseases/metabolism , CD27 Ligand/antagonists & inhibitors , Neoplasms/therapy , Tumor Necrosis Factor Receptor Superfamily, Member 7/antagonists & inhibitors , Animals , Autoimmune Diseases/therapy , CD27 Ligand/metabolism , Humans , Mice , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...