Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Clin Cancer Res ; 30(3): 532-541, 2024 02 01.
Article in English | MEDLINE | ID: mdl-37939105

ABSTRACT

PURPOSE: Eftilagimod alpha (efti), a soluble lymphocyte activation gene (LAG-3) protein and MHC class II agonist, enhances innate and adaptive immunity. Active Immunotherapy PAClitaxel (AIPAC) evaluated safety and efficacy of efti plus paclitaxel in patients with predominantly endocrine-resistant, hormone receptor-positive, HER2-negative metastatic breast cancer (ET-resistant HR+ HER2- MBC). PATIENTS AND METHODS: Women with HR+ HER2- MBC were randomized 1:1 to weekly intravenous paclitaxel (80 mg/m2) and subcutaneous efti (30 mg) or placebo every 2 weeks for six 4-week cycles, then monthly subcutaneous efti (30 mg) or placebo maintenance. Primary endpoint was progression-free survival (PFS) by blinded independent central review. Secondary endpoints included overall survival (OS), safety/tolerability, pharmacokinetics/pharmacodynamics, and quality of life. Exploratory endpoints included cellular biomarkers. RESULTS: 114 patients received efti and 112 patients received placebo. Median age was 60 years (91.6% visceral disease, 84.1% ET-resistant, 44.2% with previous CDK4/6 inhibitor treatment). Median PFS at 7.3 months was similar for efti and placebo. Median OS was not significantly improved for efti (20.4 vs. 17.5 months; HR, 0.88; P = 0.197) but became significant for predefined exploratory subgroups. EORTC QLQC30-B23 global health status was sustained for efti but deteriorated for placebo. Efti increased absolute lymphocyte, monocyte and secondary target cell (CD4, CD8) counts, plasma IFNγ and CXCL10 levels. CONCLUSIONS: Although the primary endpoint, PFS, was not met, AIPAC confirmed expected pharmacodynamic effects and demonstrated excellent safety profile for efti. OS was not significantly improved globally (2.9-month difference), but was significantly improved in exploratory biomarker subgroups, warranting further studies to clarify efti's role in patients with ET-resistant HER2- MBC.


Subject(s)
Breast Neoplasms , Female , Humans , Middle Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/pathology , Disease-Free Survival , Lymphocyte Activation , Paclitaxel , Quality of Life , Receptor, ErbB-2/metabolism
2.
Front Immunol ; 12: 634127, 2021.
Article in English | MEDLINE | ID: mdl-33828550

ABSTRACT

Sepsis is associated with a dysregulated inflammatory response to infection. Despite the activation of inflammation, an immune suppression is often observed, predisposing patients to secondary infections. Therapies directed at restoration of immunity may be considered but should be guided by the immune status of the patients. In this paper, we described the use of a high-dimensional flow cytometry (HDCyto) panel to assess the immunophenotype of patients with sepsis. We then isolated peripheral blood mononuclear cells (PBMCs) from patients with septic shock and mimicked a secondary infection by stimulating PBMCs for 4 h in vitro with lipopolysaccharide (LPS) with or without prior exposure to either IFN-γ, or LAG-3Ig. We evaluated the response by means of flow cytometry and high-resolution clustering cum differential analysis and compared the results to PBMCs from healthy donors. We observed a heterogeneous immune response in septic patients and identified two major subgroups: one characterized by hypo-responsiveness (Hypo) and another one by hyper-responsiveness (Hyper). Hypo and Hyper groups showed significant differences in the production of cytokines/chemokine and surface human leukocyte antigen-DR (HLA-DR) expression in response to LPS stimulation, which were observed across all cell types. When pre-treated with either interferon gamma (IFN-γ) or lymphocyte-activation gene 3 (LAG)-3 recombinant fusion protein (LAG-3Ig) prior to LPS stimulation, cells from the Hypo group were shown to be more responsive to both immunostimulants than cells from the Hyper group. Our results demonstrate the importance of patient stratification based on their immune status prior to any immune therapies. Once sufficiently scaled, this approach may be useful for prescribing the right immune therapy for the right patient at the right time, the key to the success of any therapy.


Subject(s)
Antigens, CD/pharmacology , Flow Cytometry , Immunophenotyping , Interferon-gamma/pharmacology , Leukocytes, Mononuclear/drug effects , Lipopolysaccharides/pharmacology , Monitoring, Immunologic , Shock, Septic/immunology , Biomarkers/blood , Case-Control Studies , Cells, Cultured , Cytokines/blood , HLA-DR Antigens/blood , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Phenotype , Predictive Value of Tests , Shock, Septic/blood , Shock, Septic/diagnosis , Workflow , Lymphocyte Activation Gene 3 Protein
3.
Eur J Immunol ; 51(2): 331-341, 2021 02.
Article in English | MEDLINE | ID: mdl-32920841

ABSTRACT

Immune checkpoint inhibitors (antibodies that block the T cell co-inhibitory receptors PD-1/PD-L1 or CTLA-4) have revolutionized the treatment of some forms of cancer. Importantly, combination approaches using drugs that target both pathways have been shown to boost the efficacy of such treatments. Subsequently, several other T cell inhibitory receptors have been identified for the development of novel immune checkpoint inhibitors. Included in this list is the co-inhibitory receptor lymphocyte activation gene-3 (LAG-3), which is upregulated on T cells extracted from tumor sites that have suppressive or exhausted phenotypes. However, the molecular rules that govern the function of LAG-3 are still not understood. Using surface plasmon resonance combined with a novel bead-based assay (AlphaScreenTM ), we demonstrate that LAG-3 can directly and specifically interact with intact human leukocyte antigen class II (HLA-II) heterodimers. Unlike the homologue CD4, which has an immeasurably weak affinity using these biophysical approaches, LAG-3 binds with low micromolar affinity. We further validated the interaction at the cell surface by staining LAG-3+ cells with pHLA-II-multimers. These data provide new insights into the mechanism by which LAG-3 initiates T cell inhibition.


Subject(s)
Antigens, CD/immunology , Costimulatory and Inhibitory T-Cell Receptors/immunology , HLA Antigens/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , CD4 Antigens/immunology , Cell Line, Tumor , Humans , Immune Checkpoint Inhibitors/immunology , Jurkat Cells , Neoplasms/immunology , Lymphocyte Activation Gene 3 Protein
4.
J Immunother Cancer ; 8(2)2020 11.
Article in English | MEDLINE | ID: mdl-33219094

ABSTRACT

BACKGROUND: To evaluate the safety, tolerability, pharmacokinetics and pharmacodynamics of eftilagimod alpha (efti), a soluble lymphocyte activation gene-3 protein, in combination with the programmed cell death-1 (PD-1) antagonist pembrolizumab. METHODS: The study was divided into two parts; parts A and B, where part A was the dose escalation part and part B was an extension part of the study. Patients with metastatic melanoma were treated with efti plus the standard dose of pembrolizumab. Blood samples were assayed to determine plasma pharmacokinetic parameters, detect efti antibody formation and determine long-lived CD8 T cell responses and associated pharmacodynamic parameters. RESULTS: Twenty-four patients with melanoma received pembrolizumab and bi-weekly subcutaneous (s.c.) injections of efti at doses 1 mg, 6 mg or 30 mg/injection for up to 6 months (part A) or 30 mg/injection for up 12 months (part B). No dose-limiting toxicities were reported and the main adverse event for efti was injection site reactions. Sustained systemic exposure to the product was obtained in all patients following s.c. injections of 30 mg dose. Treatment induced an increase in activated CD8 and CD4 T cell counts, and in some of the soluble biomarkers, particularly interferon (IFN)-γ, a Th1 signature cytokine. An overall response rate (ORR) of 33% was observed in patients partly with pembrolizumab-refractory of part A and ORR of 50% was observed in patients with PD-1 naïve of part B. CONCLUSIONS: Efti was well tolerated in combination with pembrolizumab with encouraging antitumor activity. This warrants further clinical studies of this new combination therapy combining an antigen-presenting cell activator with an immune checkpoint inhibitor.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, CD/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Lymphocyte Activation/drug effects , Melanoma/drug therapy , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, CD/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Female , Humans , Male , Melanoma/pathology , Lymphocyte Activation Gene 3 Protein
5.
J Immunol ; 204(4): 810-818, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31907283

ABSTRACT

T cells chronically stimulated with the same peptide tend to express exhaustion markers such as PD-1 or LAG-3. Deficiencies in the PD-1 and LAG-3 pathways have been linked to the development of autoimmune diseases. IMP761 is a LAG-3-specific humanized agonist Ab with immunosuppressive properties both in vitro and in vivo in an Ag-specific delayed-type hypersensitivity (DTH) model in the cynomolgus macaque (Macaca fascicularis). IMP761 inhibits TCR-mediated NFAT activation and Ag-induced human T cell proliferation and activation. In the DTH model, assessment of T cell infiltration and gene expression profile at the DTH biopsy site corresponds to immunosuppression of an Ag-induced T cell response. IMP761 is the first LAG-3-specific agonist product candidate, acting upstream on activated T cells, the root cause of self-Ag-specific T cell-induced autoimmune diseases.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antigens, CD/drug effects , Autoimmune Diseases/immunology , Immunosuppressive Agents/pharmacology , T-Lymphocytes/drug effects , Animals , Antigens, CD/immunology , Autoimmunity/drug effects , Autoimmunity/immunology , Cell Proliferation/drug effects , Humans , Hypersensitivity, Delayed/immunology , Lymphocyte Activation/drug effects , Macaca fascicularis , Male , T-Lymphocytes/immunology , Lymphocyte Activation Gene 3 Protein
6.
Future Oncol ; 15(17): 1963-1973, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30977393

ABSTRACT

Eftilagimod alpha (IMP321), a soluble dimeric recombinant form of LAG-3, is a first-in-class antigen presenting cell activator under clinical development. By stimulating dendritic cells through MHC class II molecules, IMP321 was proven to induce sustained immune responses. Combining active immunotherapy with a standard cytotoxic chemotherapy regimen represents a promising novel strategy that might lead to therapeutic improvements in metastatic breast cancer. Here, we describe the rationale and design of AIPAC (NCT02614833), a double-blind, randomized, multicenter Phase IIb study evaluating IMP321 plus paclitaxel as a first-line chemotherapy compared with paclitaxel plus placebo in hormone receptor-positive metastatic breast cancer patients. The primary end point is progression-free survival and key secondary objectives include overall survival, safety, quality of life and objective response rate.


Subject(s)
Antigens, CD/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Paclitaxel/administration & dosage , Adult , Antigens, CD/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Clinical Trials, Phase II as Topic , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Humans , Multicenter Studies as Topic , Paclitaxel/adverse effects , Placebos/administration & dosage , Placebos/adverse effects , Progression-Free Survival , Quality of Life , Randomized Controlled Trials as Topic , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Lymphocyte Activation Gene 3 Protein
7.
J Immunother Cancer ; 5: 50, 2017.
Article in English | MEDLINE | ID: mdl-28649381

ABSTRACT

This report is a summary of 'New Cancer Immunotherapy Agents in Development' program, which took place in association with the 31st Annual Meeting of the Society for Immunotherapy of Cancer (SITC), on November 9, 2016 in National Harbor, Maryland. Presenters gave brief overviews of emerging clinical and pre-clinical immune-based agents and combinations, before participating in an extended panel discussion with multidisciplinary leaders, including members of the FDA, leading academic institutions and industrial drug developers, to consider topics relevant to the future of cancer immunotherapy.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy , Neoplasms/drug therapy , Tumor Microenvironment/immunology , Cancer Vaccines/immunology , Humans , Neoplasms/immunology , Tumor Microenvironment/drug effects
8.
Clin Cancer Res ; 22(6): 1330-40, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26500235

ABSTRACT

PURPOSE: Cancer vaccines aim to generate and maintain antitumor immune responses. We designed a phase I/IIa clinical trial to test a vaccine formulation composed of Montanide ISA-51 (Incomplete Freund's Adjuvant), LAG-3Ig (IMP321, a non-Toll like Receptor agonist with adjuvant properties), and five synthetic peptides derived from tumor-associated antigens (four short 9/10-mers targeting CD8 T-cells, and one longer 15-mer targeting CD4 T-cells). Primary endpoints were safety and T-cell responses. EXPERIMENTAL DESIGN: Sixteen metastatic melanoma patients received serial vaccinations. Up to nine injections were subcutaneously administered in three cycles, each with three vaccinations every 3 weeks, with 6 to 14 weeks interval between cycles. Blood samples were collected at baseline, 1-week after the third, sixth and ninth vaccination, and 6 months after the last vaccination. Circulating T-cells were monitored by tetramer staining directly ex vivo, and by combinatorial tetramer and cytokine staining on in vitro stimulated cells. RESULTS: Side effects were mild to moderate, comparable to vaccines with Montanide alone. Specific CD8 T-cell responses to at least one peptide formulated in the vaccine preparation were found in 13 of 16 patients. However, two of the four short peptides of the vaccine formulation did not elicit CD8 T-cell responses. Specific CD4 T-cell responses were found in all 16 patients. CONCLUSIONS: We conclude that vaccination with IMP321 is a promising and safe strategy for inducing sustained immune responses, encouraging further development for cancer vaccines as components of combination therapies.


Subject(s)
Antigens, CD/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Melanoma/immunology , Melanoma/therapy , Peptides/immunology , Antigens, CD/chemistry , Antigens, Neoplasm/immunology , Biomarkers , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Combined Modality Therapy , Female , Humans , Lymphocyte Count , MART-1 Antigen/immunology , Male , Melanoma/pathology , Treatment Outcome , Vaccination , Lymphocyte Activation Gene 3 Protein
9.
Clin Cancer Res ; 21(13): 3031-40, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25688160

ABSTRACT

PURPOSE: Clear cell renal cell carcinoma (ccRCC) has shown durable responses to checkpoint blockade therapies. However, important gaps persist in the understanding of its immune microenvironment. This study aims to investigate the expression and prognostic significance of immune checkpoints in primary and metastatic ccRCC, in relation with mature dendritic cells (DC) and T-cell densities. EXPERIMENTAL DESIGN: We investigated the infiltration and the localization of CD8(+) T cells and mature DC, and the expression of immune checkpoints (PD-1, LAG-3, PD-L1, and PD-L2) in relation with prognosis, in 135 primary ccRCC tumors and 51 ccRCC lung metastases. RNA expression data for 496 primary ccRCC samples were used as confirmatory cohort. RESULTS: We identify two groups of tumors with extensive CD8(+) T-cell infiltrates. One group, characterized by high expression of immune checkpoints in the absence of fully functional mature DC, is associated with increased risk of disease progression. The second group, characterized by low expression of immune checkpoints and localization of mature DC in peritumoral immune aggregates (tertiary lymphoid structures), is associated with good prognosis. CONCLUSIONS: The expression of the immune checkpoints and the localization of DC in the tumor microenvironment modulate the clinical impact of CD8(+) T cells in ccRCC.


Subject(s)
Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Antigens, CD/metabolism , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/physiology , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/secondary , Disease-Free Survival , Humans , Kaplan-Meier Estimate , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/physiology , Multivariate Analysis , Prognosis , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Receptor/metabolism , Proportional Hazards Models , Tumor Microenvironment , Lymphocyte Activation Gene 3 Protein
10.
J Transl Med ; 12: 97, 2014 Apr 12.
Article in English | MEDLINE | ID: mdl-24726012

ABSTRACT

BACKGROUND: Immunotherapy offers a promising novel approach for the treatment of cancer and both adoptive T-cell transfer and immune modulation lead to regression of advanced melanoma. However, the potential synergy between these two strategies remains unclear. METHODS: We investigated in 12 patients with advanced stage IV melanoma the effect of multiple MART-1 analog peptide vaccinations with (n = 6) or without (n = 6) IMP321 (LAG-3Ig fusion protein) as an adjuvant in combination with lymphodepleting chemotherapy and adoptive transfer of autologous PBMCs at day (D) 0 (Trial registration No: NCT00324623). All patients were selected on the basis of ex vivo detectable MART-1-specific CD8 T-cell responses and immunized at D0, 8, 15, 22, 28, 52, and 74 post-reinfusion. RESULTS: After immunization, a significant expansion of MART-1-specific CD8 T cells was measured in 83% (n = 5/6) and 17% (n = 1/6) of patients from the IMP321 and control groups, respectively (P < 0.02). Compared to the control group, the mean fold increase of MART-1-specific CD8 T cells in the IMP321 group was respectively >2-, >4- and >6-fold higher at D15, D30 and D60 (P < 0.02). Long-lasting MART-1-specific CD8 T-cell responses were significantly associated with IMP321 (P < 0.02). At the peak of the response, MART-1-specific CD8 T cells contained higher proportions of effector (CCR7⁻ CD45RA⁺/⁻) cells in the IMP321 group (P < 0.02) and showed no sign of exhaustion (i.e. were mostly PD1⁻CD160⁻TIM3⁻LAG3⁻2B4⁺/⁻). Moreover, IMP321 was associated with a significantly reduced expansion of regulatory T cells (P < 0.04); consistently, we observed a negative correlation between the relative expansion of MART-1-specific CD8 T cells and of regulatory T cells. Finally, although there were no confirmed responses as per RECIST criteria, a transient, 30-day partial response was observed in a patient from the IMP321 group. CONCLUSIONS: Vaccination with IMP321 as an adjuvant in combination with lymphodepleting chemotherapy and adoptive transfer of autologous PBMCs induced more robust and durable cellular antitumor immune responses, supporting further development of IMP321 as an adjuvant for future immunotherapeutic strategies.


Subject(s)
Antigens, CD/administration & dosage , Lymphocyte Depletion , MART-1 Antigen/immunology , Monocytes/transplantation , Adult , Aged , Cell Transplantation , Female , Flow Cytometry , Humans , Lymphocyte Activation , Male , Middle Aged , T-Lymphocytes, Regulatory/immunology , Lymphocyte Activation Gene 3 Protein
11.
J Invest Dermatol ; 134(7): 1893-1902, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24441096

ABSTRACT

Plasmacytoid dendritic cells (pDCs) at tumor sites are often tolerogenic. Although pDCs initiate innate and adaptive immunity upon Toll-like receptor (TLR) triggering by pathogens, TLR-independent signals may be responsible for pDC activation and immune suppression in the tumor inflammatory environment. To identify molecules that are potentially involved in alternative pDC activation, we explored the expression and function of lymphocyte activation gene 3 (LAG-3) in human pDCs. In this report, we showed the expression of LAG-3 on the cell surface of a subset of circulating human pDCs. LAG-3+ pDCs exhibited a partially mature phenotype and were enriched at tumor sites in samples from melanoma patients. We found that LAG-3 interacted with major histocompatibility complex class II (MHC-II) to induce TLR-independent activation of pDCs with limited IFNα and enhanced IL-6 production. This in vitro cytokine profile of LAG-3-activated pDCs paralleled that of tumor-associated pDCs analyzed ex vivo. By confocal microscopy, LAG-3+ pDCs detected in melanoma-invaded lymph nodes (LNs) stained positive for IL-6 and preferentially localized near melanoma cells. These results suggest that LAG-3-mediated activation of pDCs takes place in vivo at tumor sites, and it is in part responsible for directing an immune-suppressive environment.


Subject(s)
Antigens, CD/immunology , Dendritic Cells/immunology , Melanoma/immunology , Skin Neoplasms/immunology , Animals , Antigens, CD/metabolism , COS Cells , Cell Line, Tumor , Cell Movement/immunology , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Chlorocebus aethiops , Dendritic Cells/pathology , Humans , Interleukin-6/immunology , Interleukin-6/metabolism , Melanoma/pathology , Monocytes/immunology , Monocytes/pathology , Skin Neoplasms/pathology , Tumor Microenvironment/immunology , Lymphocyte Activation Gene 3 Protein
12.
Oncoimmunology ; 3(11): e967146, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25941596

ABSTRACT

We have recently reported that lymphocyte activation gene-3 (LAG-3,CD223) mediates the alternative, IFNα-deficient activation of plasmacytoid dendritic cells (pDCs) at tumor sites. Our findings define a novel tumor-driven strategy that promotes immunosuppression by pDCs, and we have provided more detailed information regarding the immunomodulatory role of of LAG-3. The translational relevance of our results for the treatment of tumors and autoimmune diseases is discussed herein.

13.
J Immunol ; 186(9): 5173-83, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21441454

ABSTRACT

Melanoma is the most aggressive skin cancer in humans that often expresses MHC class II (MHC II) molecules, which could make these tumors eliminable by the immune system. However, this MHC II expression has been associated with poor prognosis, and there is a lack of immune-mediated eradication. The lymphocyte activation gene-3 (LAG-3) is a natural ligand for MHC II that is substantially expressed on melanoma-infiltrating T cells including those endowed with potent immune-suppressive activity. Based on our previous data showing the signaling capacity of MHC II in melanoma cells, we hypothesized that LAG-3 could contribute to melanoma survival through its MHC II signaling capacity in melanoma cells. In this study, we demonstrate that both soluble LAG-3 and LAG-3-transfected cells can protect MHC II-positive melanoma cells, but not MHC II-negative cells, from FAS-mediated and drug-induced apoptosis. Interaction of LAG-3 with MHC II expressed on melanoma cells upregulates both MAPK/Erk and PI3K/Akt pathways, albeit with different kinetics. Inhibition studies using specific inhibitors of both pathways provided evidence of their involvement in the LAG-3-induced protection from apoptosis. Altogether, our data suggest that the LAG-3-MHC II interaction could be viewed as a bidirectional immune escape pathway in melanoma, with direct consequences shared by both melanoma and immune cells. In the future, compounds that efficiently hinder LAG-3-MHC II interaction might be used as an adjuvant to current therapy for MHC II-positive melanoma.


Subject(s)
Antigens, CD/immunology , Apoptosis/immunology , Histocompatibility Antigens Class II/immunology , Melanoma/immunology , Skin Neoplasms/immunology , Tumor Escape/immunology , Antigens, CD/metabolism , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique , Histocompatibility Antigens Class II/metabolism , Humans , Immunoblotting , Immunohistochemistry , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma/metabolism , Melanoma/pathology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transfection , Lymphocyte Activation Gene 3 Protein
14.
J Transl Med ; 8: 71, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20653948

ABSTRACT

BACKGROUND: IMP321 is a recombinant soluble LAG-3Ig fusion protein that binds to MHC class II with high avidity and mediates APC and then antigen-experienced memory CD8+ T cell activation. We report clinical and biological results of a phase I/II in patients with metastatic breast carcinoma (MBC) receiving first-line paclitaxel weekly, 3 weeks out of 4. METHODS: MBC patients were administered one dose of IMP321 s.c. every two weeks for a total of 24 weeks (12 injections). The repeated single doses were administered the day after chemotherapy at D2 and D16 of the 28-day cycles of paclitaxel (80 mg/m2 at D1, D8 and D15, for 6 cycles). Blood samples were taken 13 days after the sixth and the twelfth IMP321 injections to determine sustained APC, NK and memory CD8 T cell responses. RESULTS: Thirty MBC patients received IMP321 in three cohorts (doses: 0.25, 1.25 and 6.25 mg). IMP321 induced both a sustained increase in the number and activation of APC (monocytes and dendritic cells) and an increase in the percentage of NK and long-lived cytotoxic effector-memory CD8 T cells. Clinical benefit was observed for 90% of patients with only 3 progressors at 6 months. Also, the objective tumor response rate of 50% compared favorably to the 25% rate reported in the historical control group. CONCLUSIONS: The absence of toxicity and the demonstration of activity strongly support the future development of this agent for clinical use in combined first-line regimens. TRIAL REGISTRATION: ClinicalTrials.gov NCT00349934.


Subject(s)
Antigens, CD/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Immunity/immunology , Immunotherapy , Paclitaxel/therapeutic use , Aged , Antibodies, Neoplasm/immunology , Antigens, CD/adverse effects , Antigens, CD/immunology , Antigens, CD/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/pathology , Cell Count , Drug Administration Schedule , Female , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Middle Aged , Monocytes/cytology , Monocytes/drug effects , Neoplasm Metastasis , Paclitaxel/adverse effects , Paclitaxel/pharmacology , Remission Induction , Treatment Outcome , Lymphocyte Activation Gene 3 Protein
15.
J Immunol ; 184(11): 6545-51, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20421648

ABSTRACT

Human natural regulatory CD4(+) T cells comprise 5-10% of peripheral CD4(+)T cells. They constitutively express the IL-2Ralpha-chain (CD25) and the nuclear transcription Foxp3. These cells are heterogeneous and contain discrete subsets with distinct phenotypes and functions. Studies in mice report that LAG-3 has a complex role in T cell homeostasis and is expressed in CD4(+)CD25(+) T regulatory cells. In this study, we explored the expression of LAG-3 in human CD4(+) T cells and found that LAG-3 identifies a discrete subset of CD4(+)CD25(high)Foxp3(+) T cells. This CD4(+)CD25(high)Foxp3(+)LAG-3(+) population is preferentially expanded in the PBMCs of patients with cancer, in lymphocytes of tumor-invaded lymph nodes and in lymphocytes infiltrating visceral metastasis. Ex vivo analysis showed that CD4(+)CD25(high)Foxp3(+)LAG-3(+) T cells are functionally active cells that release the immunosuppressive cytokines IL-10 and TGF-beta1, but not IL-2. An in vitro suppression assay using CD4(+)CD25(high)LAG-3(+) T cells sorted from in vitro expanded CD4(+)CD25(high) regulatory T cells showed that this subset of cells is endowed with potent suppressor activity that requires cell-to-cell contact. Our data show that LAG-3 defines an active CD4(+)CD25(high)Foxp3(+) regulatory T cell subset whose frequency is enhanced in the PBMCs of patients with cancer and is expanded at tumor sites.


Subject(s)
Antigens, CD/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Antigens, CD/biosynthesis , Cell Separation , Flow Cytometry , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/immunology , Humans , Lymphocyte Activation/immunology , Lymphocyte Activation Gene 3 Protein
16.
Clin Cancer Res ; 15(19): 6225-31, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19755389

ABSTRACT

PURPOSE: To evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of IMP321, a recombinant soluble LAG-3Ig fusion protein which agonizes MHC class II-driven dendritic cell activation. EXPERIMENTAL DESIGN: Patients with advanced renal cell carcinoma were treated with escalating doses of IMP321 s.c. Blood samples were assayed to determine plasma pharmacokinetic parameters, detect human anti-IMP321 antibody formation, and determine long-lived CD8 T cell responses. RESULTS: Twenty-one advanced renal cell carcinoma patients received 119 injections of IMP321 at doses ranging from 0.050 to 30 mg/injection s.c. biweekly for 6 injections. No clinically significant adverse events were observed. Good systemic exposure to the product was obtained following s.c. injections of doses above 6 mg. IMP321 induced both sustained CD8 T-cell activation and an increase in the percentage of long-lived effector-memory CD8 T cells in all patients at doses above 6 mg. Tumor growth was reduced and progression-free survival was better in those patients receiving higher doses (>6 mg) of IMP321: 7 of 8 evaluable patients treated at the higher doses experienced stable disease at 3 months compared with only 3 of 11 in the lower dose group (P = 0.015). CONCLUSION: The absence of toxicity and the demonstration of activity at doses above 6 mg warrant further disease-directed studies of IMP321 in combined regimens (e.g., chemoimmunotherapy).


Subject(s)
Antigens, CD/metabolism , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/therapy , Recombinant Fusion Proteins/pharmacokinetics , Animals , Antigens, CD/adverse effects , Antigens, CD/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Drug Administration Schedule , Drug Evaluation, Preclinical , Genes, MHC Class II/immunology , HLA-D Antigens/immunology , Humans , Immunotherapy/methods , Kidney Neoplasms/immunology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Macaca fascicularis , Maximum Tolerated Dose , Neoplasm Staging , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/therapeutic use , Tumor Cells, Cultured , Lymphocyte Activation Gene 3 Protein
18.
Clin Cancer Res ; 14(11): 3545-54, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18519788

ABSTRACT

PURPOSE: The purpose of the present study was to evaluate granulocyte macrophage colony-stimulating factor (GM-CSF)-secreting tumor cell immunotherapy, which is known to stimulate a potent and long-lasting antigen-specific immune response in combination with lymphocyte activation gene-3 fusion protein (LAG-3Ig), which has been shown to act as an adjuvant for priming T helper type 1 and cytotoxic T-cell responses. EXPERIMENTAL DESIGN: Survival and immune monitoring studies were done in the B16 melanoma model. GM-CSF-secreting tumor cell immunotherapy was administered as a single s.c. injection and LAG-3Ig was administered s.c. at the immunotherapy site. RESULTS: The studies reported here show that combining LAG-3Ig with GM-CSF-secreting tumor cell immunotherapy prolonged the survival of tumor-bearing animals compared with animals treated with either therapy alone. Prolonged survival correlated with increased numbers of systemic IFN gamma-secreting CD8+ T cells and a significantly increased infiltration of activated effector CD8+ T cells into the tumor. Moreover, an increase in antigen-specific IgG1 humoral responses was detected in serum of animals injected with the combination therapy compared with animals injected with either therapy alone. CONCLUSION: LAG-3Ig combined with a GM-CSF-secreting tumor cell immunotherapy stimulated both cellular and humoral antitumor immune responses that correlated with prolonged survival in tumor-bearing animals.


Subject(s)
Antigens, CD/therapeutic use , Immunotherapy/methods , Macrophage Colony-Stimulating Factor/therapeutic use , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Animals , Antibodies/blood , Combined Modality Therapy , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology , Lymphocyte Activation Gene 3 Protein
19.
J Immunol ; 180(6): 3782-8, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18322184

ABSTRACT

Data have been reported on the in vivo adjuvant role of soluble lymphocyte activation gene-3 (LAG-3) recombinant protein in mouse models and on its ability to support the in vitro generation of human, tumor-specific CTLs. In this study, we show that soluble human rLAG-3 protein (hLAG-3Ig) used in vitro as a single maturation agent induces phenotypic maturation of monocyte-derived dendritic cells and promoted the production of chemokines and TNF-alpha inflammatory cytokine. When given in association with optimal or suboptimal doses of CD40/CD40L, hLAG-3Ig functions as a strong costimulatory factor and induces full functional activation of monocyte-derived dendritic cells that includes the production of high level of IL-12p70. Moreover, evidence is here provided that this costimulatory function licensing dendritic cells to produce IL-12p70 is also a functional property of LAG-3 molecules when expressed in a physiological context by CD4(+) activated T cells. Altogether, these data show for the first time a role of LAG-3 in mediating dendritic cell activation when expressed on the T cell surface or released after specific Ag stimulation in the interspaces of immunological synapses.


Subject(s)
Antigens, CD/biosynthesis , Antigens, CD/genetics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lymphocyte Activation/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Amino Acid Sequence , Animals , Antigens, CD/physiology , CHO Cells , Cell Line, Tumor , Chemokines/biosynthesis , Coculture Techniques , Cricetinae , Cricetulus , Humans , Immunophenotyping , Inflammation Mediators/metabolism , Interleukin-12/metabolism , Mice , Molecular Sequence Data , Monocytes/immunology , Monocytes/metabolism , NIH 3T3 Cells , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/physiology , Lymphocyte Activation Gene 3 Protein
20.
Transplantation ; 84(11): 1500-6, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18091527

ABSTRACT

BACKGROUND: Lymphocyte-activated gene-3 (LAG-3, CD223) is upregulated during the early stages of T-cell activation and could be the target of cytotoxic antibodies for induction therapy in transplantation. METHODS: Fully vascularized heterotopic allogeneic heart transplantation was performed in rats across a full major histocompatibility complex-mismatch barrier (LEW.1W into LEW.1A). Recipients received two injections (day 0 and 3) of cytotoxic antibodies directed to the extra-loop of LAG-3 immunoglobulin (Ig)-like N-terminal domain or control antibodies. RESULTS: LAG-3 mRNA transcripts accumulated in cardiac allografts undergoing rejection, but not in peripheral lymphoid organs. Administration of anti-LAG-3 antibodies on the day of transplantation did not modify alloreactivity of T lymphocytes from the spleen and did not change the alloantibody response. However, it inhibited graft infiltration by effector mononuclear cells, reduced intragraft levels of interferon-gamma mRNA and prolonged allograft survival from 6 days in controls to a median of 27 days. Anti-LAG-3 antibodies were also active in prolonging survival when administered in a delayed manner, after rejection onset. LAG-3 being also expressed by activated regulatory T (Treg) cells, we tested the effect of anti-LAG-3 antibodies on graft acceptance after donor blood transfusions, a Treg-dependent tolerance induction model. We found that tolerance induction was prevented by anti-LAG-3 antibodies. CONCLUSIONS: Targeting LAG-3-positive cells with cytotoxic antibodies is immunosuppressive in transplantation by depleting effectors T cells and therefore may represent a treatment for rejection episodes focused only on pathogenic cells. However, it might not be compatible with tolerance-induction strategies.


Subject(s)
Antigens, CD/immunology , Antigens, CD/metabolism , Graft Rejection/immunology , Heart Transplantation , Immune Tolerance/immunology , Animals , Antibodies/immunology , Antibodies/toxicity , Antigens, CD/genetics , Blood Transfusion , Gene Expression Regulation , Graft Survival , Hemocyanins/immunology , Immunization , Lymphocytes/immunology , Lymphocytes/metabolism , Male , RNA, Messenger/genetics , Rats , Spleen/drug effects , T-Lymphocytes/immunology , Transplantation, Homologous/immunology , Lymphocyte Activation Gene 3 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...