Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 306
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Clin Immunol ; 251: 109275, 2023 06.
Article in English | MEDLINE | ID: mdl-36870379

ABSTRACT

Alternatives are urgently needed in patients with CD3- CD4+ lymphocytic-variant hypereosinophilic syndrome (L-HES) requiring high-level steroids or who are unresponsive and/or intolerant to conventional alternative therapies. We report five L-HES patients (44-66 years) with cutaneous involvement (n = 5) and persistent eosinophilia (n = 3) despite conventional therapies, who successfully received JAK inhibitors (tofacitinib n = 1, ruxolitinib n = 4). JAKi led to complete clinical remission in the first 3 months in all (with prednisone withdrawal in four). Absolute eosinophil counts normalized in cases receiving ruxolitinib, while reduction was partial under tofacitinib. After switch from tofacitinib to ruxolitinib, complete clinical response persisted despite prednisone withdrawal. The clone size remained stable in all patients. After 3-13 months of follow-up, no adverse event was reported. Prospective clinical trials are warranted to examine the use of JAKi in L-HES.


Subject(s)
Hypereosinophilic Syndrome , Humans , Prednisone/therapeutic use , Prospective Studies , CD3 Complex , Hypereosinophilic Syndrome/drug therapy , CD4-Positive T-Lymphocytes
2.
Nutrients ; 13(9)2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34578993

ABSTRACT

Eicosapentaenoic acid (20:5n-3) and docosahexaenoic acid (22:6n-3) are important for leukocyte function. This study investigated whether consuming transgenic Camelina sativa (tCSO) seed oil containing both 20:5n-3 and 22:6n-3 is as effective as fish oil (FO) for increasing the 20:5n-3 and 22:6n-3 content of leukocytes and altering mitogen-induced changes to the T cell transcriptome. Healthy adults (n = 31) consumed 450 mg/day of 20:5n-3 plus 22:6n-3 from either FO or tCSO for 8 weeks. Blood was collected before and after the intervention. 20:5n-3 and 22:6n-3 incorporation from tCSO into immune cell total lipids was comparable to FO. The relative expression of the transcriptomes of mitogen-stimulated versus unstimulated T lymphocytes in a subgroup of 16 women/test oil showed 4390 transcripts were differentially expressed at Baseline (59% up-regulated), 4769 (57% up-regulated) after FO and 3443 (38% up-regulated) after tCSO supplementation. The 20 most altered transcripts after supplementation differed between test oils. The most altered pathways were associated with cell proliferation and immune function. In conclusion, 20:5n-3 and 22:6n-3 incorporation into immune cells from tCSO was comparable to FO and can modify mitogen-induced changes in the T cell transcriptome, contingent on the lipid matrix of the oil.


Subject(s)
Brassicaceae/chemistry , Dietary Supplements , Fish Oils/pharmacology , Plant Oils/pharmacology , T-Lymphocytes/drug effects , Transcriptome/drug effects , Adolescent , Adult , Aged , CD3 Complex , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Female , Healthy Volunteers , Humans , Male , Middle Aged , T-Lymphocytes/immunology , Young Adult
3.
J Allergy Clin Immunol ; 148(4): 1081-1087.e2, 2021 10.
Article in English | MEDLINE | ID: mdl-34019913

ABSTRACT

BACKGROUND: CD56-expressing natural killer (NK) cells as well as invariant NK T (iNKT) cells have been shown to either promote or inhibit allergic immune responses. OBJECTIVE: The aim of the present study was to investigate the impact of these cells in a recently developed humanized mouse model of allergen-induced IgE-dependent gut and lung inflammation. METHODS: Nonobese diabetic-severe combined immunodeficiency γ-chain knockout mice were injected intraperitoneally with human PBMCs or CD56-depleted (CD56neg) PBMCs from highly sensitized donors with birch or grass pollen allergy together with the respective allergen or with NaCl as a control. Three weeks later, the mice were challenged with the allergen rectally and gut inflammation was monitored by video miniendoscopy and by histology. Furthermore, airway inflammation was measured after an additional intranasal allergen challenge. RESULTS: Allergen-specific human IgE in mouse sera, detectable only after coinjection of the respective allergen, was reduced in mice being injected with CD56neg PBMCs compared with in mice receiving nondepleted PBMCs. Consequently, allergen-induced IgE-dependent colitis, airway hyperreactivity, and mucus-producing goblet cells were significantly inhibited in these mice. Interestingly, reconstitution of CD56neg PBMCs with nondepleted CD56+ cells and with CD56+CD3+ iNKT cells restored gut as well as lung inflammation, whereas addition of CD3-depleted CD56+ cells did not. CONCLUSION: These results demonstrate that allergen-specific gut and lung inflammation in PBMC-engrafted humanized mice is promoted by CD56+CD3+ iNKT cells, which opens new possibilities of therapeutic intervention in allergic diseases.


Subject(s)
Colitis/immunology , Natural Killer T-Cells/immunology , Respiratory Hypersensitivity/immunology , Rhinitis, Allergic, Seasonal/immunology , Allergens/immunology , Animals , Betula/immunology , CD3 Complex/immunology , CD56 Antigen/immunology , Colitis/pathology , Colitis/physiopathology , Colon/immunology , Colon/pathology , Female , Humans , Immunoglobulin E/blood , Lung/immunology , Lung/pathology , Lung/physiopathology , Male , Mice, Transgenic , Poaceae/immunology , Pollen/immunology , Respiratory Hypersensitivity/pathology , Respiratory Hypersensitivity/physiopathology , Rhinitis, Allergic, Seasonal/pathology , Rhinitis, Allergic, Seasonal/physiopathology
4.
Immunotherapy ; 13(2): 125-141, 2021 02.
Article in English | MEDLINE | ID: mdl-33172323

ABSTRACT

Aim: We report results of a first-in-human study of pasotuxizumab, a PSMA bispecific T-cell engager (BiTE®) immune therapy mediating T-cell killing of tumor cells in patients with advanced castration-resistant prostate cancer. Patients & methods: We assessed once-daily subcutaneous (SC) pasotuxizumab. All SC patients developed antidrug antibodies; therefore, continuous intravenous (cIV) infusion was assessed. Results: A total of 47 patients received pasotuxizumab (SC: n = 31, 0.5-172 µg/d; cIV: n = 16, 5-80 µg/d). The SC maximum tolerated dose was 172.0 µg/d. A sponsor change stopped the cIV cohort early; maximum tolerated dose was not determined. PSA responders occurred (>50% PSA decline: SC, n = 9; cIV, n = 3), including two long-term responders. Conclusion: Data support pasotuxizumab safety in advanced castration-resistant prostate cancer and represent evidence of BiTE monotherapy efficacy in solid tumors. Clinical trial registration: NCT01723475 (ClinicalTrials.gov).


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents, Immunological , Prostatic Neoplasms, Castration-Resistant , Aged , Aged, 80 and over , Humans , Male , Middle Aged , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacokinetics , Antibodies, Bispecific/therapeutic use , Antigens, Surface/immunology , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/blood , CD3 Complex/immunology , Glutamate Carboxypeptidase II/immunology , Immunotherapy , Infusions, Intravenous , Injections, Subcutaneous , Maximum Tolerated Dose , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/immunology , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/therapy , Treatment Outcome
5.
Medicine (Baltimore) ; 99(45): e22859, 2020 Nov 06.
Article in English | MEDLINE | ID: mdl-33157928

ABSTRACT

BACKGROUND: This article will evaluate the effects of traditional Chinese medicine (TCM) combined with chemotherapy on the immune function and quality of life of patients with non-small cell lung cancer (NSCLC), and evaluate the published side effects. METHODS: The systematic review and meta-analysis will be conducted in accordance with the Preferred Reporting Items for Systematic Review and Meta-Analysis guidelines. The databases we will search include: PubMed, EMBASE, Cochrane Library, Web of Science, China National Knowledge Infrastructure, China Biomedicine, Wan fang Data, and Technology Periodical Database. The search date is from inception to June 30, 2020. There are no restrictions on the document language. The literatures included in this study are randomized controlled trials. The main results include ratio of CD3, CD4, CD8, CD4/CD8, NK cells, the level of IgA, IgG, IgM, and Karnofsky performance status score. The secondary result is to evaluate various side effects during treatment. We will use the Cochrane Collaboration tool to evaluate each study and use Review Manager software (RevMan, version 5.3) to merge and analyze the data. The 2 researchers will independently cross-screen the literature, extract data, and evaluate the quality. If there are differences, we will resolve them through discussion or consultation with a third reviewer. RESULTS: The results of this study will provide high-quality evidence for the effect of TCM combined with chemotherapy on the immune function and quality of life of patients with NSCLC. CONCLUSION: This article will comprehensively evaluate the effects of TCM combined with chemotherapy on the immune function and quality of life of patients with NSCLC, and provide evidence-based evidence for clinical practice. ETHICS: Since the data used in this study is based on previous trials and does not involve patient privacy, ethical approval is not required. STUDY REGISTRATION NUMBER: INPLASY202070071.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Medicine, Chinese Traditional , Quality of Life , Antineoplastic Agents/therapeutic use , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Non-Small-Cell Lung/blood , Humans , Immunoglobulin Isotypes/blood , Killer Cells, Natural/metabolism , Lung Neoplasms/blood , Meta-Analysis as Topic , Research Design , Systematic Reviews as Topic
6.
Theranostics ; 10(23): 10498-10512, 2020.
Article in English | MEDLINE | ID: mdl-32929362

ABSTRACT

Rationale: Current traditional treatment options are frequently ineffective to fight against ovarian cancer due to late diagnosis and high recurrence. Therefore, there is a vital need for the development of novel therapeutic agents. B7H3, an immune checkpoint protein, is highly expressed in various cancers, representing it a promising target for cancer immunotherapy. Although targeting B7H3 by bispecific T cell-engaging antibodies (BiTE) has achieved successes in hematological malignancies during recent years, attempts to use them for the treatment of solid cancers are less favorable, in part due to the heterogeneity of tumors. Sorafenib is an unselective inhibitor of multiple kinases currently being tested in clinical trials for several tumors, including ovarian cancer which showed limited activity and inevitable side effect for ovarian cancer treatment. However, it is able to enhance antitumor immune response, which indicates sorafenib may improve the efficiency of immunotherapy. Methods: We evaluated the expression of B7H3 in ovarian cancer using online database and validated its expression of tumor tissues by immunohistochemistry staining. Then, B7H3 expression and the effects of sorafenib on ovarian cancer cell lines were determined by flow cytometry. In addition, 2D and 3D ovarian cancer models were established to test the combined therapeutic effect in vitro. Finally, the efficiency of B7H3×CD3 BiTE alone and its combination with sorafenib were evaluated both in vitro and in vivo. Results: Our data showed that B7H3 was highly expressed in ovarian cancer compared with normal samples. Treatment with sorafenib inhibited ovarian cancer cell proliferation and induced a noticeable upregulation of B7H3 expression level. Further study suggested that B7H3×CD3 BiTE was effective in mediating T cell killing to cancer cells. Combined treatment of sorafenib and B7H3×CD3 BiTE had synergistic anti-tumor effects in ovarian cancer models. Conclusions: Overall, our study indicates that combination therapy with sorafenib and B7H3×CD3 BiTE may be a new therapeutic option for the further study of preclinical treatment of OC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , B7 Antigens/antagonists & inhibitors , Carcinoma, Ovarian Epithelial/therapy , Ovarian Neoplasms/therapy , Sorafenib/pharmacology , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7 Antigens/analysis , B7 Antigens/metabolism , CD3 Complex/antagonists & inhibitors , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/mortality , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Datasets as Topic , Drug Synergism , Female , HEK293 Cells , Humans , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Neoplasm Recurrence, Local , Ovarian Neoplasms/immunology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovary/pathology , Sorafenib/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
7.
Exp Dermatol ; 29(10): 993-1003, 2020 10.
Article in English | MEDLINE | ID: mdl-32737987

ABSTRACT

Skin models mimicking features of psoriasis-related inflammation are needed to support the development of new drugs in dermatology. Reconstructed skin models lack tissue complexity, including a fully competent skin barrier, and presence and/or diversity of immune cells. Here, we describe InflammaSkin®, a novel human Th17-driven ex vivo skin inflammation model. In this model, skin-resident T cells are in situ activated by intradermal injection of anti-CD3 and anti-CD28 antibodies and Th17 cell polarization is sustained by culture in a chemically defined medium supplemented with IL-1ß, IL-23 and TGF-ß for seven days. The acquired Th17 signature is demonstrated by the sustained secretion of IL-17A, IL-17AF, IL-17F, IL-22, IFN-γ, and to some degree IL-15 and TNF-α observed in the activated ex vivo skin inflammation model compared with the non-activated skin model control. Furthermore, expression of S100A7 and Keratin-16 by keratinocytes and loss of epidermal structure integrity occur subsequently to in situ Th17cell activation, demonstrating cellular crosstalk between Th17 cells and keratinocytes. Finally, we demonstrate the use of this model to investigate the modulation of the IL-23/IL-17 immune axis by topically applied anti-inflammatory compounds. Taken together, we show that by in situ activation of skin-resident Th17 cells, the InflammaSkin® model reproduces aspects of inflammatory responses observed in psoriatic lesions and could be used as a translational tool to assess efficacy of test compounds.


Subject(s)
Dermatitis/immunology , Lymphocyte Activation , Models, Biological , Th17 Cells/immunology , Anti-Inflammatory Agents/therapeutic use , Antibodies , Betamethasone/analogs & derivatives , Betamethasone/therapeutic use , CD28 Antigens/immunology , CD3 Complex/immunology , Cell Communication , Culture Media , Dermatitis/drug therapy , Humans , Interferon-gamma/metabolism , Interleukin-15/metabolism , Interleukin-17/metabolism , Interleukins/metabolism , Keratin-16/metabolism , Keratinocytes/metabolism , Phosphodiesterase 4 Inhibitors/therapeutic use , S100 Calcium Binding Protein A7/metabolism , Th17 Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-22
8.
Food Funct ; 11(7): 5992-6006, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32697211

ABSTRACT

Ziziphus Jujuba cv. Pozao has been consumed as a traditional fruit with regional characteristics in China for a long time; however, fewer studies on polysaccharides from Ziziphus Jujuba cv. Pozao (JP) have been documented. This study aimed to evaluate the effect of oral administration of JP on cyclophosphamide-induced ICR mice for 28 days. The results showed that oral administration of JP could significantly improve the lymphocyte proliferation in the spleen and decrease the proportion of CD3+ and CD4+ and the ratio of CD4+/CD8+ in cyclophosphamide-induced mice in a dose-dependent manner. JP treatment also increased the levels of IL-2, IL-4, IL-10, IFN-γ, and TNF-α in serum and the intestine, and the improvement effects were proportional to the dose of JP. Similarly, JP significantly increased the levels of IgA and SIgA, as well as the expressions of Claudin-1 and Occludin in the intestine. Particularly, the expressions of Claudin-1 and Occludin were the best in the M-JP group. Furthermore, JP positively regulated the gut microbiota as indicated by the enriched microbiota diversity. At the phylum level, the relative abundance of Firmicutes was significantly decreased by JP, while that of Bacteroidetes was increased by JP treatment. More importantly, the ratio of Firmicutes/Bacteroidetes was significantly increased. And a high dose of JP is the most effective. At the genus level, the abundances of the Bacteroidales-S24-7-group, Lachnospiraceae, Alloprevotella, Alistipes and Bacteroides were increased by JP treatment. These results provided evidence for the regulating effect of JP on the peripheral immunity and intestinal barrier function in cyclophosphamide-induced hypoimmune mice.


Subject(s)
Dietary Supplements , Gastrointestinal Microbiome/drug effects , Immunity/drug effects , Intestines/drug effects , Lymphocytes/metabolism , Polysaccharides/pharmacology , Ziziphus/chemistry , Animals , Bacteria/drug effects , CD3 Complex/metabolism , CD4 Antigens/metabolism , CD4-CD8 Ratio , Claudin-1/metabolism , Cyclophosphamide , Cytokines/metabolism , Fruit/chemistry , Immunoglobulin A/metabolism , Immunoglobulin A, Secretory/metabolism , Intestines/microbiology , Intestines/physiology , Male , Mice , Occludin/metabolism , Plant Extracts/pharmacology , Spleen/drug effects , Spleen/metabolism
9.
J Nat Prod ; 83(6): 1804-1809, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32539381

ABSTRACT

Hypersonins A-D (1-4), four 1,2-seco-homoadamantane type polycyclic polyprenylated acylphloroglucinols (PPAPs) possessing a new bicyclo[4.3.1]decane-3-methoxycarbonyl architecture, were obtained from Hypericum wilsonii. The structures of hypersonins A-D were identified by spectroscopic data, electronic circular dichroism comparison, and X-ray crystallographic data. Hypersonins A-D are the first seco-homoadamantane-type PPAPs with cleavage at the C-1-C-2 bond. Hypersonin A (1) showed moderate inhibitory activity to anti-CD3/anti-CD28 monoclonal antibody-induced proliferation of murine splenocytes, with an IC50 value of 8.3 ± 0.2 µM.


Subject(s)
Hypericum/chemistry , Polycyclic Compounds/pharmacology , Animals , Antibodies, Blocking , Antineoplastic Agents, Phytogenic/chemistry , CD28 Antigens/antagonists & inhibitors , CD3 Complex/antagonists & inhibitors , Cell Proliferation/drug effects , Circular Dichroism , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred BALB C , Molecular Structure , Polycyclic Compounds/chemistry , Spleen/cytology , X-Ray Diffraction
10.
J Immunother Cancer ; 8(1)2020 04.
Article in English | MEDLINE | ID: mdl-32273346

ABSTRACT

BACKGROUND: First-in-human (FIH) clinical trials require careful selection of a safe yet biologically relevant starting dose. Typically, such starting doses are selected based on toxicity studies in a pharmacologically relevant animal model. However, with the advent of target-specific and highly active immunotherapeutics, both the Food and Drug Administration and the European Medicines Agency have provided guidance that recommend determining a safe starting dose based on a minimum anticipated biological effect level (MABEL) approach. METHODS: We recently developed a T cell activating bispecific antibody that effectively treats orthotopic patient-derived malignant glioma and syngeneic glioblastoma in mice (hEGFRvIII:CD3 bi-scFv). hEGFRvIII:CD3 bi-scFv is comprized of two single chain antibody fragments (bi-scFvs) that bind mutant epidermal growth factor receptor variant III (EGFRvIII), a mutation frequently seen in malignant glioma, and human CD3ε on T cells, respectively. In order to establish a FIH dose, we used a MABEL approach to select a safe starting dose for hEGFRvIII:CD3 bi-scFv, based on a combination of in vitro data, in vivo animal studies, and theoretical human receptor occupancy modeling. RESULTS: Using the most conservative approach to the MABEL assessment, a dose of 57.4 ng hEGFRvIII:CD3 bi-scFv/kg body weight was selected as a safe starting dose for a FIH clinical study. CONCLUSIONS: The comparison of our MABEL-based starting dose to our in vivo efficacious dose and the theoretical human receptor occupancy strongly supports that our human starting dose of 57.4 ng hEGFRvIII:CD3 bi-scFv/patient kg will be safe.


Subject(s)
Antibodies, Bispecific/administration & dosage , Antibodies, Monoclonal/administration & dosage , CD3 Complex/immunology , ErbB Receptors/immunology , Glioma/drug therapy , Models, Theoretical , Animals , Apoptosis , Cell Proliferation , Computer Simulation , Dose-Response Relationship, Drug , Drug Dosage Calculations , Drug Evaluation, Preclinical , Female , Glioma/immunology , Glioma/pathology , Humans , Mice , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
JCI Insight ; 5(7)2020 04 09.
Article in English | MEDLINE | ID: mdl-32271166

ABSTRACT

Systemic cytokine release and on-target/off-tumor toxicity to normal tissues are the main adverse effects limiting the clinical utility of T cell-redirecting therapies. This study was designed to determine how binding affinity for CD3 and tumor target HER2 impact the efficacy and nonclinical safety of anti-HER2/CD3 T cell-dependent antibodies (TDBs). Affinity was found to be a major determinant for the overall tolerability. Higher affinity for CD3 associated with rapidly elevated peripheral cytokine concentrations, weight loss in mice, and poor tolerability in cynomolgus monkeys. A TDB with lower CD3 affinity was better tolerated in cynomolgus monkeys compared with a higher CD3-affinity TDB. In contrast to tolerability, T cell binding affinity had only limited impact on in vitro and in vivo antitumor activity. High affinity for HER2 was critical for the tumor-killing activity of anti-HER2/CD3 TDBs, but higher HER2 affinity also associated with a more severe toxicity profile, including cytokine release and damage to HER2-expressing tissues. The tolerability of the anti-HER2/CD3 was improved by implementing a dose-fractionation strategy. Fine-tuning the affinities for both the tumor target and CD3 is likely a valuable strategy for achieving maximal therapeutic index of CD3 bispecific antibodies.


Subject(s)
Antibodies, Bispecific/immunology , Antibody Affinity , Antineoplastic Agents, Immunological/immunology , Receptor, ErbB-2/immunology , Animals , Antibodies, Bispecific/chemistry , Antineoplastic Agents, Immunological/chemistry , CD3 Complex/chemistry , CHO Cells , Cricetulus , Drug Evaluation, Preclinical , Humans , Macaca fascicularis , Receptor, ErbB-2/chemistry
12.
Clin Cancer Res ; 26(9): 2203-2215, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31969333

ABSTRACT

PURPOSE: Multiple myeloma (MM) patients with disease refractory to all available drugs have a poor outcome, indicating the need for new agents with novel mechanisms of action. EXPERIMENTAL DESIGN: We evaluated the anti-MM activity of the fully human BCMA×CD3 bispecific antibody JNJ-7957 in cell lines and bone marrow (BM) samples. The impact of several tumor- and host-related factors on sensitivity to JNJ-7957 therapy was also evaluated. RESULTS: We show that JNJ-7957 has potent activity against 4 MM cell lines, against tumor cells in 48 of 49 BM samples obtained from MM patients, and in 5 of 6 BM samples obtained from primary plasma cell leukemia patients. JNJ-7957 activity was significantly enhanced in patients with prior daratumumab treatment, which was partially due to enhanced killing capacity of daratumumab-exposed effector cells. BCMA expression did not affect activity of JNJ-7957. High T-cell frequencies and high effector:target ratios were associated with improved JNJ-7957-mediated lysis of MM cells. The PD-1/PD-L1 axis had a modest negative impact on JNJ-7957 activity against tumor cells from daratumumab-naïve MM patients. Soluble BCMA impaired the ability of JNJ-7957 to kill MM cells, although higher concentrations were able to overcome this negative effect. CONCLUSIONS: JNJ-7957 effectively kills MM cells ex vivo, including those from heavily pretreated MM patients, whereby several components of the immunosuppressive BM microenvironment had only modest effects on its killing capacity. Our findings support the ongoing trial with JNJ-7957 as single agent and provide the preclinical rationale for evaluating JNJ-7957 in combination with daratumumab in MM.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/pharmacology , B-Cell Maturation Antigen/immunology , CD3 Complex/immunology , Multiple Myeloma/drug therapy , Antibodies, Bispecific/immunology , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents/pharmacology , Bone Marrow/pathology , Drug Evaluation, Preclinical , Drug Synergism , Drug Therapy, Combination , Humans , Immunotherapy/methods , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Tumor Cells, Cultured
13.
Mol Cancer Ther ; 18(11): 2008-2020, 2019 11.
Article in English | MEDLINE | ID: mdl-31434693

ABSTRACT

The restricted expression pattern of B-cell maturation antigen (BCMA) makes it an ideal tumor-associated antigen (TAA) for the treatment of myeloma. BCMA has been targeted by both CD3 bispecific antibody and antibody-drug conjugate (ADC) modalities, but a true comparison of modalities has yet to be performed. Here we utilized a single BCMA antibody to develop and characterize both a CD3 bispecific and 2 ADC formats (cleavable and noncleavable) and compared activity both in vitro and in vivo with the aim of generating an optimal therapeutic. Antibody affinity, but not epitope was influential in drug activity and hence a high-affinity BCMA antibody was selected. Both the bispecific and ADCs were potent in vitro and in vivo, causing dose-dependent cell killing of myeloma cell lines and tumor regression in orthotopic myeloma xenograft models. Primary patient cells were effectively lysed by both CD3 bispecific and ADCs, with the bispecific demonstrating improved potency, maximal cell killing, and consistency across patients. Safety was evaluated in cynomolgus monkey toxicity studies and both modalities were active based on on-target elimination of B lineage cells. Distinct nonclinical toxicity profiles were seen for the bispecific and ADC modalities. When taken together, results from this comparison of BCMA CD3 bispecific and ADC modalities suggest better efficacy and an improved toxicity profile might be achieved with the bispecific modality. This led to the advancement of a bispecific candidate into phase I clinical trials.


Subject(s)
Antibodies, Bispecific/administration & dosage , B-Cell Maturation Antigen/metabolism , CD3 Complex/immunology , Immunoconjugates/administration & dosage , Multiple Myeloma/drug therapy , Animals , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/pharmacology , Antibody Affinity , B-Cell Maturation Antigen/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoconjugates/adverse effects , Immunoconjugates/pharmacology , Mice , Multiple Myeloma/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
14.
J Immunol ; 203(3): 585-592, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31332079

ABSTRACT

Effector lymphocytes are multifunctional cells of the immune system that promote cytolysis of pathogen-infected cells and nascent tumors. Tumors must learn to evade effectors and employ a wide variety of mechanisms to do so. Bispecific Abs (BsAbs) are an emerging cancer immunotherapy approach seeking to re-engage either T effectors or NK cells with malignant cells. Possessing specificity for effector cells on one end and a tumor Ag on the other, these molecules work by attracting effectors to the target cell to build an immunologic synapse and induce tumor cell killing. The BsAb blinatumomab, for example, has specificity for the T cell-activating cell surface protein CD3 and the B cell Ag CD19. The only BsAb with regulatory approval currently, blinatumomab is used in the treatment of relapsed or refractory B cell acute lymphoblastic leukemia. Many additional BsAbs are in preclinical development, however, targeting many different tumor types. The variety of potential effector cells and cancer Ags, along with potential combination therapies, make BsAbs an active area of drug development. In this review, we discuss cancer recognition by the immune system and structural and mechanistic aspects of BsAbs. We summarize key steps in preclinical development and subsequent translation to medical practice. Future directions for BsAbs include combinations with a wide variety of both immunologic and nonimmunologic therapies. Defining their optimum clinical use is at early stages.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antineoplastic Agents/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , T-Lymphocytes/immunology , Antigens, CD19/immunology , Antigens, Neoplasm/immunology , CD3 Complex/immunology , Drug Evaluation, Preclinical , Humans , Immunotherapy/methods , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
15.
Scand J Immunol ; 90(5): e12808, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31322752

ABSTRACT

CD4+ T cell immunotherapy has potential for treatment in HIV-infected patients. A large number of expanded CD4+ T cells and confirmation of functional-related phenotypes are required for ensuring the successful outcomes of treatment. Freshly isolated CD4+ T cells from healthy donors were activated with anti-CD3/28-coated magnetic beads at different bead-to-cell ratios and cultured in the absence and presence of IL-2 supplementation for 3 weeks. Fold expansion, cell viability, growth kinetic and lymphocyte subset identities were determined. Data demonstrated that a 1:1 bead-to-cell ratio rendered the highest expansion of 1044-fold with 88% viability and 99.5% purity followed by the 2:1 and 0.5:1 ratios. No significant difference in proliferation and phenotypes was found between non-IL-2 and IL-2 supplementation groups. Several specific surface molecule expressions of the expanded cells including chemokine receptors, adhesion molecules, co-stimulatory molecules, activation molecules, maturation markers, cytokine receptors and other molecules were altered when compared to the unexpanded cells. This optimized expansion protocol using the 1:1 bead-to-cell ratio of anti-CD3/28-coated magnetic beads and culture condition without IL-2 supplementation provided the satisfactory yield with good reproducibility. Specific surface molecule expressions of the expanded cells presented potential roles in proliferation, differentiation, homeostasis, apoptosis and organ homing.


Subject(s)
CD28 Antigens/immunology , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , HIV Infections/therapy , Immunotherapy, Adoptive/methods , Magnetite Nanoparticles/therapeutic use , Adult , Cell Proliferation , Cells, Cultured , Coated Materials, Biocompatible , Humans , Interleukin-2/immunology , Lymphocyte Activation/immunology , Treatment Outcome
16.
World J Urol ; 37(3): 539-544, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30027406

ABSTRACT

PURPOSE: To evaluate the effect of hexanic extract of Serenoa repens (HESr) on prostatic inflammation in patients with diagnosed prostatic inflammation. METHODS: Patients with prostatic inflammation histologically confirmed by TRUS prostatic biopsy were randomized either to receive HESr (320 mg/day) or no treatment. A second biopsy was performed 6 months later according to standard clinical practice. Inflammation was assessed by the Irani's score and immunohistochemical staining using the CD3, CD4 and CD8 (for T-leucocytes), CD20 (for B-leucocytes) and CD163 (for macrophages) antibodies. RESULTS: Overall 97 patients were eligible for analysis. In the HESr group the mean inflammation grading and aggressiveness grading score significantly decreased from 1.55 and 1.55 at baseline to 0.79 (p = 0.001) and 0.87 (p = 0.001) at the second biopsy, respectively. In the control group the mean inflammation grading score was 1.44 at first biopsy and 1.23 at the second biopsy. The mean aggressiveness gradings core was 1.09 and 0.89, respectively. No statistical significance was found (p = 0.09 and p = 0.74).The mean decrease in all inflammation scores was statistically higher in the HESr patients compared to controls. The immunohistochemical staining showed a significant change in the expression of the analyzed antibodies for the HESr patients compared to the first biopsy. In the nontreatment group, no significant difference was found at the second biopsy. The change in expression of each antibody in the HESr group was statistical significant compared to control. CONCLUSIONS: HESr seems to reduce prostatic inflammation in terms of histological and immunohistochemical parameters in this specific patients population.


Subject(s)
B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Macrophages/pathology , Phytotherapy , Plant Extracts/therapeutic use , Prostate/pathology , Prostatitis/drug therapy , Serenoa , Aged , Antigens, CD/metabolism , Antigens, CD20/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biopsy , CD3 Complex/metabolism , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Hexanes , Humans , Inflammation , Macrophages/immunology , Macrophages/metabolism , Male , Middle Aged , Prostate/immunology , Prostate/metabolism , Prostatitis/immunology , Prostatitis/metabolism , Prostatitis/pathology , Receptors, Cell Surface/metabolism
17.
J Immunotoxicol ; 15(1): 119-125, 2018 12.
Article in English | MEDLINE | ID: mdl-30241454

ABSTRACT

The immunotoxic potential of drug candidates is assessed through the examination of results from a variety of studies and endpoints. While the functional assessment of CD8+ cytotoxic T-lymphocytes (CTL) is well-characterized in the clinic, the lack of a robust macaque CTL functional assay has been an important hurdle in evaluating and accurately quantifying cell-mediated CD8+ T-cell effector responses in the nonclinical setting. This paper describes the development of an assay to measure CTL activity in peripheral blood mononuclear cells (PBMC) isolated from Cynomolgus macaques. A human EGFR/CD3 Bispecific T-cell Engager (BiTE®) was used to mount a robust CD8+ T-cell response in the presence of target-expressing cells. Upon target engagement, degranulation of CD107a and production of interferon (IFN)-γ both reliably indicated a robust functional response in CD8+ T-cells. The BiTE®-mediated stimulation method proved to be favorable when compared to other methods of stimulation in the absence of target cells. These studies demonstrated acceptable longitudinal variability of the functional assay and sensitivity to dexamethasone-mediated immunosuppression. Taken together, the results indicated an assay leveraging CD3-bispecific antibodies and target-expressing cells can provide a robust approach to the in vitro or ex vivo assessment of CTL function in Cynomolgus macaques. Because the impairment of CTL activity by immunomodulators is recognized to be an important contributor to decreased antiviral defense and increased carcinogenicity risk, we believe that this novel assay to be a valuable addition to the immunotoxicology assessment of therapeutic drug candidates.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunoassay/methods , Macaca fascicularis/immunology , Animals , Antibodies, Bispecific/metabolism , CD3 Complex/immunology , Cells, Cultured , Drug Evaluation, Preclinical , ErbB Receptors/immunology , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/cytology , Lymphocyte Activation , Lysosomal-Associated Membrane Protein 1/metabolism , Observer Variation
18.
Sci Transl Med ; 10(459)2018 09 19.
Article in English | MEDLINE | ID: mdl-30232226

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) caused by a recently identified bunyavirus, SFTSV, is an emerging infectious disease with extensive geographical distribution and high mortality. Progressive viral replication and severe thrombocytopenia are key features of SFTSV infection and fatal outcome, whereas the underlying mechanisms are unknown. We revealed arginine deficiency in SFTS cases by performing metabolomics analysis on two independent patient cohorts, suggesting that arginine metabolism by nitric oxide synthase and arginase is a key pathway in SFTSV infection and consequential death. Arginine deficiency was associated with decreased intraplatelet nitric oxide (Plt-NO) concentration, platelet activation, and thrombocytopenia. An expansion of arginase-expressing granulocytic myeloid-derived suppressor cells was observed, which was related to T cell CD3-ζ chain down-regulation and virus clearance disturbance, implicating a role of arginase activity and arginine depletion in the impaired anti-SFTSV T cell function. Moreover, a comprehensive measurement of arginine bioavailability, global arginine bioavailability ratio, was shown to be a good prognostic marker for fatal prediction in early infection. A randomized controlled trial demonstrated that arginine administration was correlated with enhanced Plt-NO concentration, suppressed platelet activation, and elevated CD3-ζ chain expression and eventually associated with an accelerated virus clearance and thrombocytopenia recovery. Together, our findings revealed the arginine catabolism pathway-associated regulation of platelet homeostasis and T cell dysregulation after SFTSV infection, which not only provided a functional mechanism underlying SFTS pathogenesis but also offered an alternative therapy choice for SFTS.


Subject(s)
Arginine/deficiency , Bunyaviridae Infections/complications , Bunyaviridae Infections/immunology , Immunosuppression Therapy , Phlebovirus/physiology , Thrombocytopenia/complications , Thrombocytopenia/virology , Arginine/therapeutic use , Blood Platelets/metabolism , Bunyaviridae Infections/blood , Bunyaviridae Infections/drug therapy , CD3 Complex/metabolism , Dietary Supplements , Humans , Immunity , Metabolome , Metabolomics , Myeloid-Derived Suppressor Cells/metabolism , Nitric Oxide/metabolism , T-Lymphocytes/immunology , Thrombocytopenia/blood , Thrombocytopenia/drug therapy
19.
Acta Cir Bras ; 33(7): 556-564, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30110057

ABSTRACT

PURPOSE: To investigate the effects of baicalin on inflammatory reaction, oxidative stress and protein kinase D1 (PKD1) and nuclear factor-kappa B (NF-κB) protein expressions in severe acute pancreatitis (SAP) rats. METHODS: Sixty rats were divided into sham operation, model, and low-, medium- and high-dose baicalin group. SAP model was established in later 4 groups. The later 3 groups were injected with 0.1, 0.2 and 0.4 ml/100 g 5% baicalin injection, respectively. At 12 h, the serum SAP related indexes and inflammatory factors, peripheral blood CD3 and γδT cell percentages, wet/dry ratio and pancreas ascites volume, oxidative stress indexes and PKD1 and NF-κB protein expressions in pancreatic tissue were determined. RESULTS: Compared with model group, in high-dose baicalin group the wet/dry ratio and ascites volume, serum amylase level, phospholipase A2 activity, TNF-α, IL-1 and IL-6 levels, and pancreatic malondialdehyde level and PKD1 and NF-κB protein expression were significantly decreased (P < 0.05), and peripheral blood CD3 and γδT cell percentages and pancreatic superoxide dismutase and glutathione peroxidase levels were significantly increased (P < 0.05). CONCLUSION: Baicalin can resist the inflammatory reaction and oxidative stress, and down-regulate protein kinase D1 and nuclear factor-kappa B protein expressions, thus exerting the protective effects on severe acute pancreatitis in rats.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Flavonoids/pharmacology , NF-kappa B/metabolism , Oxidative Stress/drug effects , Pancreatitis/drug therapy , Protein Kinase C/metabolism , Amylases/blood , Amylases/drug effects , Animals , CD3 Complex/blood , CD3 Complex/drug effects , Down-Regulation/drug effects , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/metabolism , Interleukin-1/blood , Interleukin-6/blood , Malondialdehyde/metabolism , NF-kappa B/drug effects , Pancreatitis/metabolism , Protein Kinase C/drug effects , Random Allocation , Rats, Sprague-Dawley , Reproducibility of Results , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism , Treatment Outcome , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/drug effects
20.
J Autoimmun ; 94: 45-55, 2018 11.
Article in English | MEDLINE | ID: mdl-30054208

ABSTRACT

A missense C1858T single nucleotide polymorphism within PTPN22 is a strong genetic risk factor for the development of multiple autoimmune diseases. PTPN22 encodes a protein tyrosine phosphatase that negatively regulates immuno-receptor proximal Src and Syk family kinases. Notably, PTPN22 negatively regulates kinases downstream of T-cell receptor (TCR) and LFA-1, thereby setting thresholds for T-cell activation. Alterations to the quality of TCR and LFA-1 engagement at the immune synapse and the regulation of downstream signals can have profound effects on the type of effector T-cell response induced. Here we describe how IFNγ+ Th1 responses are potentiated in Ptpn22-/- T-cells and in T-cells from mice expressing Ptpn22R619W (the mouse orthologue of the human genetic variant) as they age, or following repeated immune challenge, and explore the mechanisms contributing to the expansion of Th1 cells. Specifically, we uncover two LFA-1-ICAM dependent mechanisms; one T-cell intrinsic, and one T-cell extrinsic. Firstly, we found that in vitro anti-CD3/LFA-1 induced Th1 responses were enhanced in Ptpn22-/- T-cells compared to WT, whereas anti-CD3/anti-CD28 induced IFNy responses were similar. These data were associated with an enhanced ability of Ptpn22-/- T-cells to engage ICAM-1 at the immune synapse when incubated on planar lipid bilayers, and to form conjugates with dendritic cells. Secondly, we observed a T-cell extrinsic mechanism whereby repeated stimulation of WT OT-II T-cells with LPS and OVA323-339 pulsed Ptpn22-/- bone marrow derived dendritic cells (BMDCs) was sufficient to enhance Th1 cell development compared to WT BMDCs. Furthermore, this response could be reversed by LFA-1 blockade. Our data point to two related but distinct mechanisms by which PTPN22 regulates LFA-1 dependent signals to enhance Th1 development, highlighting how perturbations to PTPN22 function over time to regulate the balance of the immune response.


Subject(s)
Arthritis, Experimental/immunology , Dendritic Cells/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , Th1 Cells/immunology , Animals , Antibodies/pharmacology , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/genetics , CD28 Antigens/immunology , CD3 Complex/antagonists & inhibitors , CD3 Complex/genetics , CD3 Complex/immunology , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Dendritic Cells/pathology , Gene Expression Regulation , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Lipid Bilayers/chemistry , Lipid Bilayers/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Function-Associated Antigen-1/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/pharmacology , Peptide Fragments/pharmacology , Polymorphism, Single Nucleotide , Protein Tyrosine Phosphatase, Non-Receptor Type 22/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Th1 Cells/drug effects , Th1 Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL