Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Cell Rep ; 27(11): 3117-3123.e5, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31189099

ABSTRACT

Agonistic antibodies targeting the tumor necrosis factor (TNF) superfamily of co-stimulatory receptors (TNFRSF) are progressing through various stages of clinical development for cancer treatment, but the desired and defining features of these agents for optimal biological activity remain controversial. One idea, based on recent studies with CD40, is that non-ligand-blocking antibodies targeting membrane-distal cysteine-rich domain 1 (CRD1) have superior agonistic activities compared with ligand-blocking antibodies targeting more membrane-proximal CRDs. Here, we determined the binding and functional characteristics of a panel of antibodies targeting CRDs 1-4 of OX40 (also known as TNFRSF4 or CD134). In striking contrast to CD40, we found that ligand-blocking CRD2-binding and membrane-proximal CRD4-binding anti-OX40 antibodies have the strongest agonistic and anti-tumor activities. These findings have important translational implications and further highlight that the relationship between epitope specificity and agonistic activity will be an important issue to resolve on a case-by-case basis when optimizing antibodies targeting different co-stimulatory tumor necrosis factor receptors (TNFRs).


Subject(s)
Antibodies, Monoclonal/immunology , Immunotherapy/methods , Neoplasms, Experimental/therapy , OX40 Ligand/immunology , Receptors, OX40/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Epitopes/chemistry , Epitopes/immunology , Humans , Jurkat Cells , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , OX40 Ligand/chemistry , Rats , Rats, Inbred Lew , Receptors, OX40/chemistry
2.
J Neuroimmune Pharmacol ; 11(1): 73-83, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26260924

ABSTRACT

Non-human primate models of human disease have an important role in the translation of a new scientific finding in lower species into an effective treatment. In this study, we tested a new therapeutic antibody against the IL-7 receptor α chain (CD127), which in a C57BL/6 mouse model of experimental autoimmune encephalomyelitis (EAE) ameliorates disease, demonstrating an important pathogenic function of IL-7. We observed that while the treatment was effective in 100 % of the mice, it was only partially effective in the EAE model in common marmosets (Callithrix jacchus), a small-bodied Neotropical primate. EAE was induced in seven female marmoset twins and treatment with the anti-CD127 mAb or PBS as control was started 21 days after immunization followed by weekly intravenous administration. The anti-CD127 mAb caused functional blockade of IL-7 signaling through its receptor as shown by reduced phosphorylation of STAT5 in lymphocytes upon stimulation with IL-7. Group-wise analysis showed no significant effects on the clinical course and neuropathology. However, paired twin analysis revealed a delayed disease onset in three twins, which were high responders to the immunization. In addition, we observed markedly opposite effects of the antibody on pathological changes in the spinal cord in high versus low responder twins. In conclusion, promising clinical effect of CD127 blockade observed in a standard inbred/SPF mouse EAE model could only be partially replicated in an outbred/non-SPF non-human primate EAE model. Only in high responders to the immunization we found a positive response to the treatment. The mechanism underpinning this dichotomous response will be discussed.


Subject(s)
Antibodies, Monoclonal/pharmacology , Encephalomyelitis, Autoimmune, Experimental/pathology , Interleukin-7 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-7/immunology , Animals , Callithrix , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Flow Cytometry , Immunohistochemistry , Polymerase Chain Reaction
3.
Cancer Immunol Res ; 3(2): 149-60, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25387892

ABSTRACT

Immunotherapies targeting the programmed death 1 (PD-1) coinhibitory receptor have shown great promise for a subset of patients with cancer. However, robust and safe combination therapies are still needed to bring the benefit of cancer immunotherapy to broader patient populations. To search for an optimal strategy of combinatorial immunotherapy, we have compared the antitumor activity of the anti-4-1BB/anti-PD-1 combination with that of the anti-PD-1/anti-LAG-3 combination in the poorly immunogenic B16F10 melanoma model. Pronounced tumor inhibition occurred only in animals receiving anti-PD-1 and anti-4-1BB concomitantly, while combining anti-PD-1 with anti-LAG-3 led to a modest degree of tumor suppression. The activity of the anti-4-1BB/anti-PD-1 combination was dependent on IFNγ and CD8(+) T cells. Both 4-1BB and PD-1 proteins were elevated on the surface of CD8(+) T cells by anti-4-1BB/anti-PD-1 cotreatment. In the tumor microenvironment, an effective antitumor immune response was induced as indicated by the increased CD8(+)/Treg ratio and the enrichment of genes such as Cd3e, Cd8a, Ifng, and Eomes. In the spleen, the combination treatment shaped the immune system to an effector/memory phenotype and increased the overall activity of tumor-specific CD8(+) CTLs, reflecting a long-lasting systemic antitumor response. Furthermore, combination treatment in C57BL/6 mice showed no additional safety signals, and only minimally increased severity of the known toxicity relative to 4-1BB agonist alone. Therefore, in the absence of any cancer vaccine, anti-4-1BB/anti-PD-1 combination therapy is sufficient to elicit a robust antitumor effector/memory T-cell response in an aggressive tumor model and is therefore a candidate for combination trials in patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/immunology , CD8-Positive T-Lymphocytes/drug effects , Melanoma, Experimental/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cytotoxicity, Immunologic/immunology , Female , Immunologic Memory/immunology , Immunophenotyping/methods , Immunotherapy/methods , Interferon-gamma/immunology , Liver/enzymology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Tumor Microenvironment/immunology , Lymphocyte Activation Gene 3 Protein
4.
Proc Natl Acad Sci U S A ; 109(31): 12674-9, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22733769

ABSTRACT

Genetic variation in the IL-7 receptor-α (IL-7R) gene is associated with susceptibility to human type 1 diabetes (T1D). Here we investigate the therapeutic efficacy and mechanism of IL-7Rα antibody in a mouse model of T1D. IL-7Rα antibody induces durable, complete remission in newly onset diabetic mice after only two to three injections. IL-7 increases, whereas IL-7Rα antibody therapy reduces, the IFN-γ-producing CD4(+) (T(H)1) and IFN-γ-producing CD8(+) T cells. Conversely, IL-7 decreases and IL-7Rα antibody enhances the inhibitory receptor Programmed Death 1 (PD-1) expression in the effector T cells. Programmed Death 1 blockade reversed the immune tolerance mediated by the IL-7Rα antibody therapy. Furthermore, IL-7Rα antibody therapy increases the frequency of regulatory T cells without affecting their suppressor activity. The durable efficacy and the multipronged tolerogenic mechanisms of IL-7Rα antibody therapy suggest a unique disease-modifying approach to T1D.


Subject(s)
Antibodies/pharmacology , Diabetes Mellitus, Type 1/immunology , Interleukin-7/immunology , Receptors, Interleukin-7/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Antibodies/immunology , Antigens, Differentiation/immunology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Disease Models, Animal , Female , Humans , Interferon-gamma/immunology , Mice , Mice, Inbred NOD , Programmed Cell Death 1 Receptor , Receptors, Interleukin-7/immunology , T-Lymphocytes, Regulatory/pathology , Th1 Cells/pathology
5.
Sci Transl Med ; 3(93): 93ra68, 2011 Jul 27.
Article in English | MEDLINE | ID: mdl-21795588

ABSTRACT

The interleukin-7 receptor α chain (IL-7Rα) gene was identified as a top non-major histocompatibility complex-linked risk locus for multiple sclerosis (MS). Recently, we showed that a T helper 1 (T(H)1)-driven, but not a T(H)17-driven, form of MS exhibited a good clinical response to interferon-ß (IFN-ß) therapy. We now demonstrate that high serum levels of IL-7, particularly when paired with low levels of IL-17F, predict responsiveness to IFN-ß and hence a T(H)1-driven subtype of MS. We also show that although IL-7 signaling is neither necessary nor sufficient for the induction or expansion of T(H)17 cells, IL-7 can greatly enhance both human and mouse T(H)1 cell differentiation. IL-7 alone is sufficient to induce human T(H)1 differentiation in the absence of IL-12 or other cytokines. Furthermore, targeting IL-7/IL-7Rα is beneficial in experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. Mice treated with IL-7Rα-blocking antibodies before or after onset of paralysis exhibited reduced clinical signs of EAE, with reduction in peripheral naïve and activated T cells, whereas central memory T, regulatory T, B, and natural killer cell populations were largely spared. IL-7Rα antibody treatment markedly reduced lymphocyte infiltration into the central nervous system in mice with EAE. Thus, a serum profile of high IL-7 may signify a T(H)1-driven form of MS and may predict outcome in MS patients undergoing IFN-ß therapy. Blockade of IL-7 and the IL-7Rα pathway may have therapeutic potential in MS and other autoimmune diseases.


Subject(s)
Interferon-beta/therapeutic use , Interleukin-7/blood , Interleukin-7/immunology , Multiple Sclerosis/blood , Multiple Sclerosis/drug therapy , Th1 Cells/cytology , Th1 Cells/immunology , Animals , Antibodies/pharmacology , Antibody Specificity/drug effects , Cell Differentiation/drug effects , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Immunologic Memory/drug effects , Interferon-beta/immunology , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Receptors, Interleukin-7/antagonists & inhibitors , Th1 Cells/drug effects , Treatment Outcome
6.
Cancer Immunol Immunother ; 59(9): 1313-23, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20499060

ABSTRACT

A critical factor in clinical development of cancer immunotherapies is the identification of tumor-associated antigens that may be related to immunotherapy potency. In this study, protein microarrays containing >8,000 human proteins were screened with serum from prostate cancer patients (N = 13) before and after treatment with a granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting whole cell immunotherapy. Thirty-three proteins were identified that displayed significantly elevated (P

Subject(s)
Antibodies/blood , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , HSP90 Heat-Shock Proteins/biosynthesis , Immunotherapy, Adoptive , Nuclear Proteins/biosynthesis , Prostatic Neoplasms/immunology , Antibodies/immunology , Biomarkers, Pharmacological , Biomarkers, Tumor/immunology , Clinical Trials as Topic , Disease Progression , Galectins/immunology , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/immunology , High-Throughput Screening Assays , Humans , Male , Neoplasm Metastasis , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Prognosis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Protein Array Analysis , Retrospective Studies , Survival Analysis , Treatment Outcome
7.
Mol Ther ; 15(6): 1153-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17375065

ABSTRACT

Monoclonal antibody (mAb) delivery by gene transfer in vivo may be an attractive alternative to current mAb therapies for applications that require long-term therapy. This article describes a transfer system that allows inducible high-level expression of unmodified mAbs in vivo. A recombinant adeno-associated viral (rAAV) vector is used that comprises an expression cassette consisting of a dimerizer-regulated promoter that drives expression of the antibody heavy and light chains linked by a 2A self-processing peptide and a furin cleavage site. Following intravenous injection of the rAAV vector, serum mAb levels >1 mg/ml were attained by administration of the inducer, rapamycin. Antibody expression could be rapidly shut off by discontinuing treatment with rapamycin. By optimizing the furin cleavage sequence, this system generated native antibody in vivo, decreasing the likelihood of a host immune response to foreign sequences. In summary, this optimized mAb expression system allows regulated high-level expression of native full-length mAbs in vivo and may offer a new opportunity for delivery of therapeutic mAbs in the clinic.


Subject(s)
Antibodies, Monoclonal/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Amino Acid Sequence , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/metabolism , Binding Sites , Blotting, Western , CHO Cells , Cell Line , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Enzyme-Linked Immunosorbent Assay , Female , Furin/metabolism , Gene Expression/drug effects , Genetic Vectors/administration & dosage , Humans , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Nude , Molecular Sequence Data , Sirolimus/pharmacology
8.
Cancer Res ; 65(15): 6901-9, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16061674

ABSTRACT

The presence of metastases in regional lymph nodes is a strong indicator of poor patient survival in many types of cancer. It has recently been shown that the lymphangiogenic growth factor, vascular endothelial growth factor-C (VEGF-C), and its receptor, VEGF receptor-3 (VEGFR3), may play a pivotal role in the promotion of metastasis to regional lymph nodes. In this study, human prostate and melanoma tumor models that preferentially metastasize to the lymph nodes following s.c. tumor cell implantation were established from lymph node metastases via in vivo selection. Melanoma tumor cell sublines established from lymph node metastasis express higher amounts of VEGF-C than the parental tumor cells. The inhibition of tumor-derived VEGF-C with a soluble VEGFR3 decoy receptor, sVEGFR3-Fc, expressed via a recombinant adeno-associated viral vector, potently blocks tumor-associated lymphangiogenesis and tumor metastasis to the lymph nodes, when the treatment was initiated before the tumor implantation. In addition, sVEGFR3-Fc serum levels required for efficient blockade of lymph node metastases are strictly dependent on the VEGF-C levels generated by the primary tumor. Recombinant adeno-associated virus-mediated gene transfer of sVEGFR3-Fc may represent a feasible therapeutic strategy for blockade of lymphogenous metastasis.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Neoplasms/therapy , Vascular Endothelial Growth Factor Receptor-3/genetics , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Cell Line, Tumor , Female , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Melanoma/genetics , Melanoma/pathology , Melanoma/therapy , Mice , Mice, Nude , Neoplasms/genetics , Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Vascular Endothelial Growth Factor Receptor-3/biosynthesis , Vascular Endothelial Growth Factor Receptor-3/blood
9.
Nat Biotechnol ; 23(5): 584-90, 2005 May.
Article in English | MEDLINE | ID: mdl-15834403

ABSTRACT

Therapeutic monoclonal antibodies (mAbs) are currently being developed for the treatment of cancer and other diseases. Despite clinical success, widespread application of mAb therapies may be limited by manufacturing capabilities. In this paper, we describe a mAb delivery system that allows continuous production of a full-length antibody at high-concentrations in vivo after gene transfer. The mAb is expressed from a single open reading frame by linking the heavy and light chains with a 2A self-processing peptide derived from the foot-and-mouth disease virus. Using this expression system, we generated a recombinant adeno-associated virus vector encoding the VEGFR2-neutralizing mAb DC101 (rAAV8-DC101). A single dose of rAAV8-DC101 resulted in long-term expression of >1,000 microg/ml of DC101 in mice, demonstrating significant anti-tumor efficacy. This report describes the first feasible gene therapy approach for stable delivery of mAbs at therapeutic levels, which may serve as an attractive alternative to direct injection of mAbs.


Subject(s)
Adenoviridae/genetics , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/therapeutic use , Antibody Formation/genetics , Genetic Therapy/methods , Kidney/metabolism , Neoplasms/therapy , Viral Proteins/metabolism , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/isolation & purification , Cell Line , Gene Transfer Techniques , Humans , Hybridomas , Kidney/immunology , Mice , Neoplasms/genetics , Neoplasms/immunology , Protein Engineering/methods , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/immunology , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL