Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Med ; 27(10): 1797-1805, 2021 10.
Article in English | MEDLINE | ID: mdl-34642489

ABSTRACT

Chimeric antigen receptor (CAR) T cells targeting CD19 or CD22 have shown remarkable activity in B cell acute lymphoblastic leukemia (B-ALL). The major cause of treatment failure is antigen downregulation or loss. Dual antigen targeting could potentially prevent this, but the clinical safety and efficacy of CAR T cells targeting both CD19 and CD22 remain unclear. We conducted a phase 1 trial in pediatric and young adult patients with relapsed or refractory B-ALL (n = 15) to test AUTO3, autologous transduced T cells expressing both anti-CD19 and anti-CD22 CARs (AMELIA trial, EUDRA CT 2016-004680-39). The primary endpoints were the incidence of grade 3-5 toxicity in the dose-limiting toxicity period and the frequency of dose-limiting toxicities. Secondary endpoints included the rate of morphological remission (complete response or complete response with incomplete bone marrow recovery) with minimal residual disease-negative response, as well as the frequency and severity of adverse events, expansion and persistence of AUTO3, duration of B cell aplasia, and overall and event-free survival. The study endpoints were met. AUTO3 showed a favorable safety profile, with no dose-limiting toxicities or cases of AUTO3-related severe cytokine release syndrome or neurotoxicity reported. At 1 month after treatment the remission rate (that is, complete response or complete response with incomplete bone marrow recovery) was 86% (13 of 15 patients). The 1 year overall and event-free survival rates were 60% and 32%, respectively. Relapses were probably due to limited long-term AUTO3 persistence. Strategies to improve CAR T cell persistence are needed to fully realize the potential of dual targeting CAR T cell therapy in B-ALL.


Subject(s)
Antigens, CD19/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Chimeric Antigen/administration & dosage , Sialic Acid Binding Ig-like Lectin 2/genetics , Adolescent , Adult , Antigens, CD19/immunology , Child , Child, Preschool , Female , Humans , Immunotherapy/adverse effects , Immunotherapy/trends , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/trends , Infant , Male , Pediatrics , Progression-Free Survival , Receptors, Chimeric Antigen/immunology , Sialic Acid Binding Ig-like Lectin 2/immunology , Young Adult
2.
Nat Cancer ; 1(12): 1153-1166, 2020 12.
Article in English | MEDLINE | ID: mdl-33644766

ABSTRACT

Intratumoral regulatory T cell (Treg) abundance associates with diminished anti-tumor immunity and poor prognosis in human cancers. Recent work demonstrates that CD25, the high affinity receptor subunit for IL-2, is a selective target for Treg depletion in mouse and human malignancies; however, anti-human CD25 antibodies have failed to deliver clinical responses against solid tumors due to bystander IL-2 receptor signaling blockade on effector T cells, which limits their anti-tumor activity. Here we demonstrate potent single-agent activity of anti-CD25 antibodies optimized to deplete Tregs whilst preserving IL-2-STAT5 signaling on effector T cells, and demonstrate synergy with immune checkpoint blockade in vivo. Pre-clinical evaluation of an anti-human CD25 (RG6292) antibody with equivalent features demonstrates, in both non-human primates and humanized mouse models, efficient Treg depletion with no overt immune-related toxicities. Our data supports the clinical development of RG6292 and evaluation of novel combination therapies incorporating non-IL-2 blocking anti-CD25 antibodies in clinical studies.


Subject(s)
Interleukin-2 , Neoplasms , Animals , Antibodies, Monoclonal/pharmacology , Interleukin-2/pharmacology , Mice , Signal Transduction , T-Lymphocytes, Regulatory
3.
Cancer Cell ; 33(4): 649-663.e4, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29576375

ABSTRACT

With the use of a mouse model expressing human Fc-gamma receptors (FcγRs), we demonstrated that antibodies with isotypes equivalent to ipilimumab and tremelimumab mediate intra-tumoral regulatory T (Treg) cell depletion in vivo, increasing the CD8+ to Treg cell ratio and promoting tumor rejection. Antibodies with improved FcγR binding profiles drove superior anti-tumor responses and survival. In patients with advanced melanoma, response to ipilimumab was associated with the CD16a-V158F high affinity polymorphism. Such activity only appeared relevant in the context of inflamed tumors, explaining the modest response rates observed in the clinical setting. Our data suggest that the activity of anti-CTLA-4 in inflamed tumors may be improved through enhancement of FcγR binding, whereas poorly infiltrated tumors will likely require combination approaches.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Melanoma/drug therapy , Polymorphism, Single Nucleotide , Receptors, IgG/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents, Immunological/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Female , Humans , Ipilimumab/administration & dosage , Ipilimumab/pharmacology , Melanoma/genetics , Melanoma/immunology , Mice , Receptors, IgG/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
4.
Immunity ; 46(4): 577-586, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28410988

ABSTRACT

CD25 is expressed at high levels on regulatory T (Treg) cells and was initially proposed as a target for cancer immunotherapy. However, anti-CD25 antibodies have displayed limited activity against established tumors. We demonstrated that CD25 expression is largely restricted to tumor-infiltrating Treg cells in mice and humans. While existing anti-CD25 antibodies were observed to deplete Treg cells in the periphery, upregulation of the inhibitory Fc gamma receptor (FcγR) IIb at the tumor site prevented intra-tumoral Treg cell depletion, which may underlie the lack of anti-tumor activity previously observed in pre-clinical models. Use of an anti-CD25 antibody with enhanced binding to activating FcγRs led to effective depletion of tumor-infiltrating Treg cells, increased effector to Treg cell ratios, and improved control of established tumors. Combination with anti-programmed cell death protein-1 antibodies promoted complete tumor rejection, demonstrating the relevance of CD25 as a therapeutic target and promising substrate for future combination approaches in immune-oncology.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin Fc Fragments/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Flow Cytometry , Humans , Immunotherapy/methods , K562 Cells , Kaplan-Meier Estimate , Lymphocyte Depletion , Mice , Neoplasms/pathology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Protein Binding/immunology , Receptors, IgG/immunology , Receptors, IgG/metabolism , T-Lymphocytes, Regulatory/metabolism
5.
Mol Cell ; 65(4): 730-742.e5, 2017 Feb 16.
Article in English | MEDLINE | ID: mdl-28212753

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is known for specifically killing cancer cells, whereas in resistant cancers, TRAIL/TRAIL-R can promote metastasis via Rac1 and PI3K. It remains unknown, however, whether and to what extent TRAIL/TRAIL-R signaling in cancer cells can affect the immune microenvironment. Here we show that TRAIL-triggered cytokine secretion from TRAIL-resistant cancer cells is FADD dependent and identify the TRAIL-induced secretome to drive monocyte polarization to myeloid-derived suppressor cells (MDSCs) and M2-like macrophages. TRAIL-R suppression in tumor cells impaired CCL2 production and diminished both lung MDSC presence and tumor growth. In accordance, the receptor of CCL2, CCR2, is required to facilitate increased MDSC presence and tumor growth. Finally, TRAIL and CCL2 are co-regulated with MDSC/M2 markers in lung adenocarcinoma patients. Collectively, endogenous TRAIL/TRAIL-R-mediated CCL2 secretion promotes accumulation of tumor-supportive immune cells in the cancer microenvironment, thereby revealing a tumor-supportive immune-modulatory role of the TRAIL/TRAIL-R system in cancer biology.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cytokines/metabolism , Lung Neoplasms/metabolism , Macrophages/metabolism , Receptors, CCR2/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Microenvironment , A549 Cells , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 8/genetics , Caspase 8/metabolism , Cell Proliferation , Chemokine CCL2/metabolism , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Female , HCT116 Cells , HeLa Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Macrophages/immunology , Macrophages/pathology , Mice, Inbred C57BL , Mice, SCID , Phenotype , RNA Interference , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction , Time Factors , Transfection , Tumor Burden
6.
Oncoimmunology ; 5(7): e1186323, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27622041

ABSTRACT

In November 2015, the CALYM Carnot Institute held a 2-d workshop to discuss the current and future development of immunomodulatory antibodies for the treatment of lymphoma. Highlights from the workshop are presented in this article.

7.
Trends Immunol ; 36(7): 388-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26091729

ABSTRACT

Anti-tumor monoclonal antibodies eliminate tumor cells through different mechanisms including antibody-mediated cell cytotoxicity and generation of long-term anti-tumor T cell responses. In a recent publication, DiLillo and Ravetch demonstrate how this vaccinal effect is mediated by engagement of Fcγ receptors expressed on antigen-presenting cells.


Subject(s)
Antibodies, Monoclonal/immunology , Neoplasms/immunology , Receptors, IgG/immunology , Animals , Humans
8.
Blood ; 124(11): 1748-64, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25006125

ABSTRACT

Acute inflammation is traditionally characterized by polymorphonuclear leukocytes (PMN) influx followed by phagocytosing macrophage (Mφs) that clear injurious stimuli leading to resolution and tissue homeostasis. However, using the peritoneal cavity, we found that although innate immune-mediated responses to low-dose zymosan or bacteria resolve within days, these stimuli, but not hyperinflammatory stimuli, trigger a previously overlooked second wave of leukocyte influx into tissues that persists for weeks. These cells comprise distinct populations of tissue-resident Mφs (resMφs), Ly6c(hi) monocyte-derived Mφs (moMφs), monocyte-derived dendritic cells (moDCs), and myeloid-derived suppressor cells (MDSCs). Postresolution mononuclear phagocytes were observed alongside lymph node expansion and increased numbers of blood and peritoneal memory T and B lymphocytes. The resMφs and moMφs triggered FoxP3 expression within CD4 cells, whereas moDCs drive T-cell proliferation. The resMφs preferentially clear apoptotic PMNs and migrate to lymph nodes to bring about their contraction in an inducible nitric oxide synthase-dependent manner. Finally, moMφs remain in tissues for months postresolution, alongside altered numbers of T cells collectively dictating the magnitude of subsequent acute inflammatory reactions. These data challenge the prevailing idea that resolution leads back to homeostasis and asserts that resolution acts as a bridge between innate and adaptive immunity, as well as tissue reprogramming.


Subject(s)
Adaptive Immunity/physiology , Phagocytosis/physiology , Adaptive Immunity/drug effects , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Movement/drug effects , Cell Movement/genetics , Cell Movement/immunology , Cell Proliferation/drug effects , Dendritic Cells/cytology , Dendritic Cells/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Mice , Monocytes/cytology , Monocytes/immunology , Phagocytosis/drug effects , Zymosan/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL