Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Discov ; 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38563906

ABSTRACT

IL-2 signals pleiotropically on diverse cell types, some of which contribute to therapeutic activity against tumors, while others drive undesired activity, such as immunosuppression or toxicity. We explored the theory that targeting of IL-2 to CD8+ T cells, which are key anti-tumor effectors, could enhance its therapeutic index. To this aim, we developed AB248, CD8 cis-targeted IL-2 that demonstrates over 500-fold preference for CD8+ T cells over NK and Treg cells, which may contribute to toxicity and immunosuppression, respectively. AB248 recapitulated IL-2's effects on CD8+ T cells in vitro and induced selective expansion of CD8+ T cells in primates. In mice, an AB248 surrogate demonstrated superior anti-tumor activity and enhanced tolerability as compared to an untargeted IL-2RBy agonist. Efficacy was associated with expansion and phenotypic enhancement of tumor-infiltrating CD8+ T cells, including the emergence of a "better effector" population. These data support the potential utility of AB248 in clinical settings.

2.
Sci Transl Med ; 16(729): eadi1572, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38198572

ABSTRACT

CD8+ T cells are key antiviral effectors against hepatitis B virus (HBV), yet their number and function can be compromised in chronic infections. Preclinical HBV models displaying CD8+ T cell dysfunction showed that interleukin-2 (IL-2)-based treatment, unlike programmed cell death ligand 1 (PD-L1) checkpoint blockade, could reverse this defect, suggesting its therapeutic potential against HBV. However, IL-2's effectiveness is hindered by its pleiotropic nature, because its receptor is found on various immune cells, including regulatory T (Treg) cells and natural killer (NK) cells, which can counteract antiviral responses or contribute to toxicity, respectively. To address this, we developed a cis-targeted CD8-IL2 fusion protein, aiming to selectively stimulate dysfunctional CD8+ T cells in chronic HBV. In a mouse model, CD8-IL2 boosted the number of HBV-reactive CD8+ T cells in the liver without substantially altering Treg or NK cell counts. These expanded CD8+ T cells exhibited increased interferon-γ and granzyme B production, demonstrating enhanced functionality. CD8-IL2 treatment resulted in substantial antiviral effects, evidenced by marked reductions in viremia and antigenemia and HBV core antigen-positive hepatocytes. In contrast, an untargeted CTRL-IL2 led to predominant NK cell expansion, minimal CD8+ T cell expansion, negligible changes in effector molecules, and minimal antiviral activity. Human CD8-IL2 trials in cynomolgus monkeys mirrored these results, achieving a roughly 20-fold increase in peripheral blood CD8+ T cells without affecting NK or Treg cell numbers. These data support the development of CD8-IL2 as a therapy for chronic HBV infection.


Subject(s)
Hepatitis B, Chronic , Interleukin-2 , Humans , Animals , Mice , Hepatitis B virus , CD8-Positive T-Lymphocytes , Hepatitis B, Chronic/drug therapy , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use
3.
Mol Ther ; 28(3): 889-900, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31981494

ABSTRACT

FLT3 (FMS-like tyrosine kinase 3), expressed on the surface of acute myeloid leukemia (AML) blasts, is a promising AML target, given its role in the development and progression of leukemia, and its limited expression in tissues outside the hematopoietic system. Small molecule FLT3 kinase inhibitors have been developed, but despite having clinical efficacy, they are effective only on a subset of patients and associated with high risk of relapse. A durable therapy that can target a wider population of AML patients is needed. Here, we developed an anti-FLT3-CD3 immunoglobulin G (IgG)-based bispecific antibody (7370) with a high affinity for FLT3 and a long half-life, to target FLT3-expressing AML blasts, irrespective of FLT3 mutational status. We demonstrated that 7370 has picomolar potency against AML cell lines in vitro and in vivo. 7370 was also capable of activating T cells from AML patients, redirecting their cytotoxic activity against autologous blasts at low effector-to-target (E:T) ratio. Additionally, under our dosing regimen, 7370 was well tolerated and exhibited potent efficacy in cynomolgus monkeys by inducing complete but reversible depletion of peripheral FLT3+ dendritic cells (DCs) and bone marrow FLT3+ stem cells and progenitors. Overall, our results support further clinical development of 7370 to broadly target AML patients.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/pharmacology , CD3 Complex/antagonists & inhibitors , Hematopoiesis/drug effects , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/therapeutic use , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/therapeutic use , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , CD3 Complex/chemistry , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Immunoglobulin G/pharmacology , Immunophenotyping , Leukemia, Myeloid, Acute , Lymphocyte Depletion , Macaca fascicularis , Mice , Models, Molecular , Protein Domains/drug effects , Structure-Activity Relationship , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays , fms-Like Tyrosine Kinase 3/chemistry
4.
Nat Commun ; 9(1): 4679, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30410017

ABSTRACT

4-1BB (CD137, TNFRSF9) is an inducible costimulatory receptor expressed on activated T cells. Clinical trials of two agonist antibodies, utomilumab (PF-05082566) and urelumab (BMS-663513), are ongoing in multiple cancer indications, and both antibodies demonstrate distinct activities in the clinic. To understand these differences, we solved structures of the human 4-1BB/4-1BBL complex, the 4-1BBL trimer alone, and 4-1BB bound to utomilumab or urelumab. The 4-1BB/4-1BBL complex displays a unique interaction between receptor and ligand when compared with other TNF family members. Furthermore, our ligand-only structure differs from previously published data. Utomilumab, a ligand-blocking antibody, binds 4-1BB between CRDs 3 and 4. In contrast, urelumab binds 4-1BB CRD-1, away from the ligand binding site. Finally, cell-based assays demonstrate utomilumab is a milder agonist than urelumab. Collectively, our data provide a deeper understanding of the 4-1BB signaling complex, providing a template for future development of next generation 4-1BB targeted biologics.


Subject(s)
4-1BB Ligand/chemistry , 4-1BB Ligand/metabolism , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/chemistry , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Antibodies, Monoclonal, Humanized , Binding Sites , HEK293 Cells , Humans , Jurkat Cells , Models, Molecular , Protein Domains
5.
J Pharmacol Exp Ther ; 340(2): 228-36, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22019884

ABSTRACT

Proprotein convertase substilisin/kexin type 9 (PCSK9) promotes the degradation of low-density lipoprotein (LDL) receptor (LDLR) and thereby increases serum LDL-cholesterol (LDL-C). We have developed a humanized monoclonal antibody that recognizes the LDLR binding domain of PCSK9. This antibody, J16, and its precursor mouse antibody, J10, potently inhibit PCSK9 binding to the LDLR extracellular domain and PCSK9-mediated down-regulation of LDLR in vitro. In vivo, J10 effectively reduces serum cholesterol in C57BL/6 mice fed normal chow. J16 reduces LDL-C in healthy and diet-induced hypercholesterolemic cynomologous monkeys, but does not significantly affect high-density lipoprotein-cholesterol. Furthermore, J16 greatly lowered LDL-C in hypercholesterolemic monkeys treated with the HMG-CoA reductase inhibitor simvastatin. Our data demonstrate that anti-PCSK9 antibody is a promising LDL-C-lowering agent that is both efficacious and potentially additive to current therapies.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Cholesterol, LDL/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Primates , Proprotein Convertases/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Catalytic Domain/immunology , Cell Line, Tumor , Cholesterol/blood , Cholesterol, Dietary/administration & dosage , Cholesterol, Dietary/pharmacology , Cholesterol, HDL/blood , Cholesterol, HDL/drug effects , Cholesterol, LDL/blood , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Dose-Response Relationship, Drug , Drug Therapy, Combination/methods , Epitopes/immunology , Female , Fluorobenzenes/pharmacology , Fluorobenzenes/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hypercholesterolemia/blood , Hypercholesterolemia/chemically induced , Hypercholesterolemia/drug therapy , Liver/drug effects , Liver/metabolism , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Proprotein Convertase 9 , Proprotein Convertases/immunology , Proprotein Convertases/pharmacology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Receptors, LDL/metabolism , Rosuvastatin Calcium , Serine Endopeptidases/blood , Serine Endopeptidases/immunology , Serine Endopeptidases/pharmacology , Simvastatin/pharmacology , Simvastatin/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...