Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cancer Immunol Immunother ; 71(1): 97-109, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34028568

ABSTRACT

Cyclophosphamide plus fludarabine (C/F) are currently used to improve the expansion and effectiveness of adoptive cell therapy (ACT). However, these chemotherapeutics cause pan-leukopenia and adverse events, suggesting that safer and more effective conditioning treatments are needed to improve ACT outcomes. Previously, we reported that varlilumab, a CD27-targeting antibody, mediates Treg -preferential T cell depletion, CD8-T cell dominant costimulation, and systemic immune activation in hCD27 transgenic mice and cancer patients. We reasoned that the activities induced by varlilumab may provide an effective conditioning regimen for ACT. Varlilumab pretreatment of hCD27 +/+mCD27 - /- mice resulted in prominent proliferation of transferred T cells isolated from wild-type mice. These studies uncovered a critical role for CD27 signaling for the expansion of transferred T cells, as transfer of T cells from CD27 deficient mice or treatment with a CD70 blocking antibody greatly reduced their proliferation. In this model, varlilumab depletes endogenous hCD27+/+ T cells and blocks their subsequent access to CD70, allowing for more CD70 costimulation available to the mCD27 +/+ transferred T cells. CD27-targeted depletion led to a greater expansion of transferred T cells compared to C/F conditioning and resulted in longer median survival and more cures than C/F conditioning in the E.G7 tumor model receiving OT-I cell therapy. We propose that translation of this work could be achieved through engineering of T cells for ACT to abrogate varlilumab binding but preserve CD70 ligation. Thus, varlilumab could be an option to chemotherapy as a conditioning regimen for ACT.


Subject(s)
Adoptive Transfer , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/chemistry , Neoplasms/therapy , T-Lymphocytes/cytology , Tumor Necrosis Factor Receptor Superfamily, Member 7/chemistry , Animals , CD27 Ligand/immunology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Cell Proliferation , Immune System , Immunotherapy , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms/metabolism , Signal Transduction , Transplantation Conditioning , Treatment Outcome
2.
Nat Commun ; 11(1): 5415, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33110069

ABSTRACT

The ability of cancer cells to ensure T-cell exclusion from the tumor microenvironment is a significant mechanism of resistance to anti-PD-1/PD-L1 therapy. Evidence indicates crucial roles of Batf3-dependent conventional type-1 dendritic cells (cDC1s) for inducing antitumor T-cell immunity; however, strategies to maximize cDC1 engagement remain elusive. Here, using multiple orthotopic tumor mouse models resistant to anti-PD-L1-therapy, we are testing the hypothesis that in situ induction and activation of tumor-residing cDC1s overcomes poor T-cell infiltration. In situ immunomodulation with Flt3L, radiotherapy, and TLR3/CD40 stimulation induces an influx of stem-like Tcf1+ Slamf6+ CD8+ T cells, triggers regression not only of primary, but also untreated distant tumors, and renders tumors responsive to anti-PD-L1 therapy. Furthermore, serial in situ immunomodulation (ISIM) reshapes repertoires of intratumoral T cells, overcomes acquired resistance to anti-PD-L1 therapy, and establishes tumor-specific immunological memory. These findings provide new insights into cDC1 biology as a critical determinant to overcome mechanisms of intratumoral T-cell exclusion.


Subject(s)
Antibodies/administration & dosage , Dendritic Cells/immunology , Neoplasms/drug therapy , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cell Line, Tumor , Drug Resistance , Female , Humans , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Tumor Microenvironment
3.
J Immunol ; 205(7): 1867-1877, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32848036

ABSTRACT

In vivo expansion of adoptively transferred CD8+ T cells is a critical determinant of successful adoptive T cell therapy. Emerging evidence indicates Batf3-dependent conventional type 1 dendritic cells (cDC1s) rarely found within the tumor myeloid compartment are crucial for effector T cell recruitment to the tumor microenvironment. However, the role of cDC1s in expansion of tumor-specific CD8+ T cells remains unclear. In this article, we addressed the role of cDC1s and their costimulatory molecules, CD40, CD70, and CD80/CD86, in expansion and antitumor efficacy of adoptively transferred in vitro-primed CD8+ T cells recognizing nonmutated tumor-associated self-antigens. We found that TLR/CD40-mediated expansion and antitumor efficacy of adoptively transferred tumor-specific CD8+ T cells were abrogated in Batf3-/- mice. Further mechanistic studies using mixed bone marrow chimeric mice identified that CD40 and CD70 but not CD80/CD86 signaling in cDC1s played a critical role in expansion and antitumor efficacy of adoptively transferred CD8+ T cells. Moreover, induction and activation of cDC1s by administration of FMS-like tyrosine kinase 3 ligand (Flt3L) and TLR/CD40 agonists augmented expansion of adoptively transferred CD8+ T cells, delayed tumor growth, and improved survival. These findings reveal a key role for CD40 and CD70 signaling in cDC1s and have major implications for the design of new vaccination strategies with adoptive T cell therapy.


Subject(s)
CD27 Ligand/metabolism , CD40 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Melanoma/immunology , Animals , Antigens, Neoplasm/immunology , Basic-Leucine Zipper Transcription Factors , CD8-Positive T-Lymphocytes/transplantation , Cells, Cultured , Cytokines/metabolism , Lymphocyte Activation , Melanoma, Experimental , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins , Signal Transduction , Th1 Cells/immunology , Th2 Cells/immunology
4.
Cancer Immunol Immunother ; 69(10): 2125-2137, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32451681

ABSTRACT

CD27 is a costimulatory molecule that provides a complementary target to the PD-1/PD-L1 checkpoint axis on T cells. Combining a CD27 agonist antibody with PD-1/PD-L1 blockade has shown synergistic antitumor activity in preclinical models, which led to clinical studies of the combination in cancer patients. We theorized that coupling CD27 costimulation with PD-1/PD-L1 blockade in a bispecific antibody (BsAb) may provide greater immune activating properties than combining the individual mAbs due to enhanced CD27 activation by cross-linking through PD-L1 and Fc receptors. To test this approach, we developed CDX-527, a tetravalent PD-L1xCD27 IgG1-scFv BsAb. CDX-527 potently inhibits PD-1 signaling and induces CD27-mediated T cell costimulation through PD-L1 cross-linking. In mixed lymphocyte reaction assays, CDX-527 is more potent than the combination of the parental antibodies, suggesting that cross-linking through both Fc receptors and PD-L1 results in enhanced CD27 agonist activity. CDX-527 was shown to mediate effector function against tumor cells overexpressing either CD27 or PD-L1. In human CD27 transgenic mice, we observed that antigen-specific T cell responses to a vaccine are greatly enhanced with a surrogate PD-L1xCD27 BsAb. Furthermore, the BsAb exhibits greater antitumor activity than the combination of the parental antibodies in a syngeneic lymphoma model. A pilot study of CDX-527 in cynomolgus macaques confirmed a mAb-like pharmacokinetic profile without noted toxicities. These studies demonstrate that CDX-527 effectively combines PD-1 blockade and CD27 costimulation into one molecule that is more potent than combination of the parental antibodies providing the rationale to advance this BsAb toward clinical studies in cancer patients.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibody Formation , Immunotherapy/methods , Lymphoma, B-Cell/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 7/antagonists & inhibitors , Animals , Antibodies, Bispecific/chemistry , Humans , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Macaca fascicularis , Male , Mice , Mice, Transgenic
5.
Cancer Immunol Immunother ; 68(2): 233-245, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30382327

ABSTRACT

Limitations of immunotherapy include poorly functioning events early in the immune response cycle, such as efficient antigen presentation and T cell priming. CD40 signaling in dendritic cells leads to upregulation of cell surface costimulatory and MHC molecules and the generation of cytokines, which promotes effective priming of CD8+ effector T cells while minimizing T cell anergy and the generation of regulatory T cells. This naturally occurs through interaction with CD40 ligand (CD40L) expressed on CD4+ T-helper cells. CD40 signaling can also be achieved using specific antibodies, leading to several agonist CD40 antibodies entering clinical development. Our approach to select a CD40 agonist antibody was to define a balanced profile between sufficiently strong immune stimulation and the untoward effects of systemic immune activation. CDX-1140 is a human IgG2 antibody that activates DCs and B cells and drives NFkB stimulation in a CD40-expressing reporter cell line. These activities are Fc-independent and are maintained using an F(ab')2 fragment of the antibody. CDX-1140 binds outside of the CD40L binding site, and addition of recombinant CD40L greatly enhances DC and B activation by CDX-1140, suggesting that CDX-1140 may act synergistically with naturally expressed CD40L. CDX-1140 also has both direct and immune-mediated anti-tumor activity in xenograft models. CDX-1140 does not promote cytokine production in whole blood assays and has good pharmacodynamic and safety profiles in cynomolgus macaques. These data support the potential of CDX-1140 as part of a cancer therapy regimen, and a phase 1 trial has recently commenced.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD40 Antigens/agonists , Immunotherapy/methods , Neoplasms/therapy , Animals , Antibodies, Monoclonal/immunology , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD40 Ligand/immunology , CD40 Ligand/metabolism , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , HEK293 Cells , Humans , Macaca fascicularis , Mice, SCID , Mice, Transgenic , Neoplasms/immunology , Neoplasms/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
6.
J Immunol ; 199(12): 4110-4123, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29109120

ABSTRACT

CD27, a member of the TNFR superfamily, is constitutively expressed in most T cells and plays crucial roles in T cell effector functions. The costimulation and antitumor activity of CD27 agonistic Abs have been well documented in mouse models. Clinical testing of a human IgG1 anti-CD27 Ab, varlilumab (clone 1F5), is ongoing in cancer patients. In this study, we set out to further understand CD27 as an immunomodulatory target and to address the mechanism of antitumor efficacy using different IgG isotypes of 1F5 in human CD27-transgenic mice. 1F5mIgG1, the only isotype engaging inhibitory FcγRIIB expressed in B cells, elicited the most potent and broad immune response, but terminal differentiation, exhaustion, and apoptosis in the activated effector T cells were inevitable. Accordingly, this isotype was the most effective in eradicating BCL1 lymphoma but had limited efficacy in s.c. tumors. Conversely, 1F5mIgG2a, which interacts with cells expressing activating FcγRs, led to moderate immune activation, as well as to prominent reduction in the number and suppressive activity of regulatory T cells. These combined mechanisms imparted potent antitumor activity to 1F5mIgG2a, particularly against the s.c. tumors. 1F5hIgG1, varlilumab, showed balanced agonistic activity that was prominent at lower doses and depleting activity that was greater at higher doses. 1F5hIgG1 had good antitumor activity in all tumor models tested. Thus, both agonist and depleting properties contribute to the antitumor efficacy of CD27-targeted immunotherapy, and modulation of these activities in patients may be achieved by varying the dose and regimen.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Lymphocyte Depletion , Neoplasms, Experimental/drug therapy , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents, Immunological/immunology , Apoptosis , CD27 Ligand/immunology , Drug Screening Assays, Antitumor , Female , Humans , Immunoglobulin Isotypes/immunology , Immunoglobulin Isotypes/therapeutic use , Immunologic Memory , Immunotherapy , Lymphoma, B-Cell/drug therapy , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mutation, Missense , Receptors, IgG/immunology , Receptors, IgG/metabolism , Specific Pathogen-Free Organisms , Tumor Microenvironment , Tumor Necrosis Factor Receptor Superfamily, Member 7/agonists , Tumor Necrosis Factor Receptor Superfamily, Member 7/antagonists & inhibitors
7.
Mol Cancer Ther ; 15(12): 2946-2954, 2016 12.
Article in English | MEDLINE | ID: mdl-27671527

ABSTRACT

T-cell immunoglobulin and mucin domain 1 (TIM-1) is a type I transmembrane protein that was originally described as kidney injury molecule 1 (KIM-1) due to its elevated expression in kidney and urine after renal injury. TIM-1 expression is also upregulated in several human cancers, most notably in renal and ovarian carcinomas, but has very restricted expression in healthy tissues, thus representing a promising target for antibody-mediated therapy. To this end, we have developed a fully human monoclonal IgG1 antibody specific for the extracellular domain of TIM-1. This antibody was shown to bind purified recombinant chimeric TIM-1-Fc protein and TIM-1 expressed on a variety of transformed cell lines, including Caki-1 (human renal clear cell carcinoma), IGROV-1 (human ovarian adenocarcinoma), and A549 (human lung carcinoma). Internalization studies using confocal microscopy revealed the antibody was rapidly internalized by cells in vitro, and internalization was confirmed by quantitative imaging flow cytometry. An antibody-drug conjugate (ADC) was produced with the anti-TIM-1 antibody covalently linked to the potent cytotoxin, monomethyl auristatin E (MMAE), and designated CDX-014. The ADC was shown to exhibit in vitro cytostatic or cytotoxic activity against a variety of TIM-1-expressing cell lines, but not on TIM-1-negative cell lines. Using the Caki-1, IGROV-1, and A549 xenograft mouse models, CDX-014 showed significant antitumor activity in a clinically relevant dose range. Safety evaluation in nonhuman primates has demonstrated a good profile and led to the initiation of clinical studies of CDX-014 in renal cell carcinoma and potentially other TIM-1-expressing tumors. Mol Cancer Ther; 15(12); 2946-54. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hepatitis A Virus Cellular Receptor 1/genetics , Immunoconjugates/pharmacology , Lung Neoplasms/genetics , Ovarian Neoplasms/genetics , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Design , Female , Hepatitis A Virus Cellular Receptor 1/chemistry , Hepatitis A Virus Cellular Receptor 1/metabolism , Humans , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Macaca fascicularis , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Protein Interaction Domains and Motifs/immunology , Xenograft Model Antitumor Assays
8.
J Immunother Cancer ; 3: 37, 2015.
Article in English | MEDLINE | ID: mdl-26500773

ABSTRACT

BACKGROUND: Clinical targeting of TNFR family of receptors (CD40, CD134 and CD137) with immunostimulatory monoclonal antibodies has been successful in cancer immunotherapy. However, targeting of CD27 with a mAb is a relatively new approach to provide costimulation of immune cells undergoing activation. Thus, activation of human CD27 (TNFRSF7) with a monoclonal antibody (varlilumab) has previously been demonstrated to result in T cell activation and anti-tumor activity in preclinical models, and is currently in early phase clinical trials in patients with advanced malignancies. In this study we used an in vitro system using human peripheral blood T cells to characterize the varlilumab-mediated costimulatory effects in combination with TCR stimulation in terms of phenotypic, transcriptional and functionality changes. METHODS: T cells were isolated from normal volunteer PBMCs using magnetic bead isolation kits and stimulated in vitro with plate bound anti-CD3 Ab (OKT3) and varlilumab or control Ab for 72 h. Activation profiles were monitored by ELISA or Luminex-based testing cytokine/chemokine releases, cell surface phenotyping for costimulatory and coinhibitory markers and CFSE dye dilution by proliferating T cells and Tregs. Changes in gene expression and transcriptome analysis of varlilumab-stimulated T cells was carried on Agilent Human whole genome microarray datasets using a suite of statistical and bioinformatic software tools. RESULTS: Costimulation of T cells with varlilumab required continuous TCR signaling as pre-activated T cells were unable to produce cytokines with CD27 signaling alone. Analysis of T cell subsets further revealed that memory CD4+ and CD8+ T cells were specifically activated with a bias toward CD8+ T lymphocyte proliferation. Activation was accompanied by upregulated cell surface expression of costimulatory [4-1BB, OX40, GITR and ICOS] and coinhibitory [PD-1] molecules. Importantly, varlilumab costimulation did not activate purified Tregs as measured by cytokine production, proliferation and suppression of dividing non-Treg T cells. Analysis of changes in gene expression during varlilumab stimulation of T cells revealed modulation of pro-inflammatory signatures consistent with cellular activation and proliferation, with the IL-2 pathway showing the highest frequency of gene modulation. CONCLUSIONS: Altogether, the data reveal the requirements and T cell subtype-specific effects of CD27 costimulation, and helps select relevant biomarkers for studying the effects of varlilumab in patients.

9.
Am J Reprod Immunol ; 73(3): 232-41, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25046333

ABSTRACT

PROBLEM: Functional complement activity is routinely measured utilizing rabbit antibody-sensitized sheep erythrocytes. Due to complement inhibitor expression on erythrocytes, the development of an alternative method to measure complement function in sheep serum was required. METHOD OF STUDY: Several species of target erythrocyte and sensitizing antibody were investigated for improved measurement of complement function testing. RESULTS AND CONCLUSION: Guinea pig erythrocytes were identified as the optimal target, although sensitizing them with rabbit antiguinea pig erythrocyte antibody did not enhance the lysis by maternal sheep serum. In contrast, preterm neonatal sheep serum was unable to efficiently lyse guinea pig erythrocytes unless pre-sensitized with antibody. Further investigation revealed that maternal serum contained high levels of antibodies that cross-reacted with guinea pig and rabbit erythrocytes, while no cross-reacting antierythrocyte antibodies were found in preterm neonatal serum. Therefore, unlike primates, rabbits, and guinea pigs, no transplacental transfer of maternal IgG to foetal sheep occurs. Use of exogenous complement regulators is often used to dissect the contribution of complement to disease pathogenesis; however, we found that while full-length soluble human complement receptor 1 (sCR1, CDX-1135) was able to inhibit lysis of guinea pig erythrocytes by human and rat serum, no inhibition of sheep serum could be observed. Investigation of complement contribution to disease pathogenesis in the future will require the identification of an inhibitor that is effective against sheep complement.


Subject(s)
Complement System Proteins/analysis , Sheep/blood , Age Factors , Animals , Animals, Newborn , Antibodies, Heterophile/immunology , Chickens , Cross Reactions , Erythrocytes , Female , Fetal Blood/immunology , Fetus/immunology , Guinea Pigs , Hemolysis , Humans , Peptide Fragments/pharmacology , Pregnancy , Rabbits , Rats , Receptors, Complement/immunology , Recombinant Proteins/pharmacology , Species Specificity
10.
Cell Mol Immunol ; 12(6): 719-28, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25345808

ABSTRACT

Previous studies have documented that selective delivery of protein antigens to cells expressing mannose receptor (MR) can lead to enhanced immune responses. We postulated that agents that influenced the MR expression level, and the activation and migration status of MR-expressing antigen presenting cells, would modulate immune responses to MR-targeted vaccines. To address this question, we investigated the effect of clinically used adjuvants in human MR transgenic (hMR-Tg) mice immunized with an MR-targeting cancer vaccine composed of the human anti-MR monoclonal antibody B11 fused with the oncofetal protein, human chorionic gonadotropin beta chain (hCGß), and referred to as B11-hCGß. We found that humoral responses to low doses of B11-hCGß could be enhanced by prior administration of GM-CSF, which upregulated MR expression in vivo. However, co-administration of the Toll-like receptor (TLR) agonists, poly-ICLC and/or CpG with B11-hCGß was required to elicit Th1 immunity, as measured by antigen-specific T-cell production of IFN-γ. The TLR agonists were shown to increase the number of vaccine-containing cells in the draining lymph nodes of immunized hMR-Tg mice. In particular, with B11-hCGß and poly-ICLC, a dramatic increase in vaccine-positive cells was observed in the T-cell areas of the lymph nodes, compared to the vaccine alone or combined with GM-CSF. Importantly, the absence of the TLR agonists during the priming immunization led to antigen-specific tolerance. Therefore, this study provides insight into the mechanisms by which adjuvants can augment immune responses to B11-hCGß and have implications for the rationale design of clinical studies combining MR-targeted vaccination with TLR agonists.


Subject(s)
Antigen-Presenting Cells/drug effects , Cancer Vaccines/genetics , Carboxymethylcellulose Sodium/analogs & derivatives , Lectins, C-Type/genetics , Mannose-Binding Lectins/genetics , Oligodeoxyribonucleotides/pharmacology , Poly I-C/pharmacology , Polylysine/analogs & derivatives , Receptors, Cell Surface/genetics , T-Lymphocytes/drug effects , Toll-Like Receptors/agonists , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Carboxymethylcellulose Sodium/pharmacology , Chorionic Gonadotropin, beta Subunit, Human/administration & dosage , Chorionic Gonadotropin, beta Subunit, Human/genetics , Chorionic Gonadotropin, beta Subunit, Human/immunology , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunity, Cellular/drug effects , Interferon-gamma/genetics , Interferon-gamma/immunology , Lectins, C-Type/immunology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Count , Mannose Receptor , Mannose-Binding Lectins/immunology , Mice , Mice, Transgenic , Polylysine/pharmacology , Receptors, Cell Surface/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Th1 Cells/cytology , Th1 Cells/drug effects , Th1 Cells/immunology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
11.
Biol Blood Marrow Transplant ; 20(3): 309-13, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24365795

ABSTRACT

Hematopoietic stem cell (HSC) transplantation has curative potential for patients with hematological malignancies. Clinically, HSCs derived from mobilized peripheral blood are used more frequently than bone marrow. However, current standard mobilizing agents yield grafts that may not contain sufficient HSCs. Here, using murine models, we discovered that FLT3L synergized with plerixafor to mobilize phenotypically defined HSCs and their combination (FP) was superior to granulocyte colony-stimulating factor (G-CSF) alone or in combination with plerixafor (GP). Additionally, FP mobilized more regulatory T cells, natural killer cells, and plasmacytoid dendritic cells compared with G-CSF alone or GP. Both syngeneic and allogeneic grafts mobilized by FP led to long-term survival in transplanted mice. Collectively, FP represents a promising novel and potent mobilization regimen with potential clinical application in both the autologous and allogeneic transplantation settings.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/mortality , Heterocyclic Compounds/pharmacology , Membrane Proteins/pharmacology , Animals , Benzylamines , Cyclams , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/transplantation , Drug Combinations , Drug Synergism , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Injections, Intraperitoneal , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Mice , Mice, Inbred BALB C , Recombinant Proteins/pharmacology , Survival Analysis , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Transplantation, Autologous , Transplantation, Homologous , Whole-Body Irradiation
12.
J Am Soc Nephrol ; 24(11): 1820-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23907509

ABSTRACT

Dense deposit disease (DDD) and C3 glomerulonephritis (C3GN) are widely recognized subtypes of C3 glomerulopathy. These ultra-rare renal diseases are characterized by fluid-phase dysregulation of the alternative complement pathway that leads to deposition of complement proteins in the renal glomerulus. Disease triggers are unknown and because targeted treatments are lacking, progress to end stage renal failure is a common final outcome. We studied soluble CR1, a potent regulator of complement activity, to test whether it restores complement regulation in C3 glomerulopathy. In vitro studies using sera from patients with DDD showed that soluble CR1 prevents dysregulation of the alternative pathway C3 convertase, even in the presence of C3 nephritic factors. In mice deficient in complement factor H and transgenic for human CR1, soluble CR1 therapy stopped alternative pathway activation, resulting in normalization of serum C3 levels and clearance of iC3b from glomerular basement membranes. Short-term use of soluble CR1 in a pediatric patient with end stage renal failure demonstrated its safety and ability to normalize activity of the terminal complement pathway. Overall, these data indicate that soluble CR1 re-establishes regulation of the alternative complement pathway and provide support for a limited trial to evaluate soluble CR1 as a treatment for DDD and C3GN.


Subject(s)
Complement C3/analysis , Glomerulonephritis, Membranoproliferative/drug therapy , Receptors, Complement 3b/therapeutic use , Animals , Child , Complement Factor H/physiology , Complement Pathway, Alternative , Glomerulonephritis, Membranoproliferative/immunology , Humans , Mice
13.
Mol Immunol ; 56(1-2): 91-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23685261

ABSTRACT

Preeclampsia is a major obstetric problem defined by new-onset hypertension and proteinuria associated with compromised placental perfusion. Although activation of the complement system is increased in preeclampsia compared to normal pregnancy, it remains unclear whether excess complement activation is a cause or consequence of placental ischemia. Therefore, we hypothesized that complement activation is critical for placental ischemia-induced hypertension. We employed the reduced utero-placental perfusion pressure (RUPP) model of placental ischemia in the rat to induce hypertension in the third trimester and evaluated the effect of inhibiting complement activation with a soluble recombinant form of an endogenous complement regulator, human complement receptor 1 (sCR1; CDX-1135). On day 14 of a 21-day gestation, rats received either RUPP or Sham surgery and 15 mg/kg/day sCR1 or saline intravenously on days 14-18. Circulating complement component 3 decreased and complement activation product C3a increased in RUPP vs. Sham (p<0.05), indicating complement activation had occurred. Mean arterial pressure (MAP) measured on day 19 increased in RUPP vs. Sham rats (109.8±2.8 mmHg vs. 93.6±1.6 mmHg). Treatment with sCR1 significantly reduced elevated MAP in RUPP rats (98.4±3.6 mmHg, p<0.05) and reduced C3a production. Vascular endothelial growth factor (VEGF) decreased in RUPP compared to Sham rats, and the decrease in VEGF was not affected by sCR1 treatment. Thus, these studies have identified a mechanistic link between complement activation and the pregnancy complication of hypertension apart from free plasma VEGF and have identified complement inhibition as a potential treatment strategy for placental ischemia-induced hypertension in preeclampsia.


Subject(s)
Complement Activation/physiology , Hypertension/physiopathology , Ischemia/physiopathology , Placenta/physiopathology , Animals , Arterial Pressure/drug effects , Carotid Arteries/drug effects , Carotid Arteries/physiopathology , Complement Activation/drug effects , Complement C3/metabolism , Dose-Response Relationship, Drug , Female , Humans , Hypertension/blood , Hypertension/metabolism , In Vitro Techniques , Male , Nitroprusside/pharmacology , Placenta/blood supply , Placenta/metabolism , Pre-Eclampsia/blood , Pre-Eclampsia/metabolism , Pre-Eclampsia/physiopathology , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Complement/administration & dosage , Time Factors , Uterus/blood supply , Uterus/metabolism , Uterus/physiopathology , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/blood , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology
14.
Hum Vaccin ; 5(2): 79-84, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18670196

ABSTRACT

Cholesteryl ester transfer protein (CETP) is a plasma glycoprotein that facilitates the transfer of neutral lipids and phospholipids between lipoproteins and contributes to the regulation of the plasma concentration of high density lipoprotein cholesterol (HDL-C). Vaccines have been developed that elicit antibodies that bind to and reduce the lipid transfer function of CETP as a way to increase the plasma concentration of HDL-C and prevent or treat atherosclerosis. This study assessed the immunogenicity of two vaccine peptides. The first, CETi-1, is a dimerized synthetic peptide, including residues 461-476 of human CETP and residues 830-843 of tetanus toxoid, TT(830-843). The second, PADRE-CETP, is a monomeric peptide, in which a PADRE T cell epitope (aK-Cha-VAAWTLKAa) replaces the TT(830-843) T cell epitope of CETi-1. Both peptides were formulated with aluminum-containing adjuvants (Alhydrogel), and tested in mice and rabbits with or without the co-administration of the investigational TLR9 agonist VaxImmune (CPG 7909). In both mice and rabbits, the vaccine peptide utilizing the PADRE T cell epitope elicited stronger anti-CETP antibody responses than the CETi-1 vaccine. Also, co-administration of VaxImmune enhanced the anti-CETP antibody responses to both vaccines. Isotype analysis of the murine anti-CETP antibody response to both vaccines demonstrated a switch from IgG1 to IgG2a upon co-administration of VaxImmune. We conclude that (1) the PADRE T cell epitope is more potent than the TT(830-843) epitope in providing help for the anti-CETP antibody response; and (2) co-administration of VaxImmune with either vaccine increased immunogenicity as measured by antibody response.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Atherosclerosis/prevention & control , Atherosclerosis/therapy , Cholesterol Ester Transfer Proteins/immunology , Oligodeoxyribonucleotides/administration & dosage , Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/pharmacology , Animals , Antibodies/blood , Female , Humans , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/pharmacology , Rabbits , Vaccines, Subunit/immunology
15.
J Mol Biol ; 375(1): 102-18, 2008 Jan 04.
Article in English | MEDLINE | ID: mdl-18028942

ABSTRACT

Human complement receptor type 1 (CR1, CD35) is a type I membrane-bound glycoprotein that belongs to the regulators of complement activity (RCA) family. The extra-cellular component of CR1 is comprised of 30 short complement regulator (SCR) domains, whereas complement receptor type 2 (CR2) has 15 SCR domains and factor H (FH) has 20 SCR domains. The domain arrangement of a soluble form of CR1 (sCR1) was studied by X-ray scattering and analytical ultracentrifugation. The radius of gyration R(G) of sCR1 of 13.4(+/-1.1) nm is not much greater than those for CR2 and FH, and its R(G)/R(0) anisotropy ratio is 3.76, compared to ratios of 3.67 for FH and 4.1 for CR2. Unlike CR2, but similar to FH, two cross-sectional R(G) ranges were identified that gave R(XS) values of 4.7(+/-0.2) nm and 1.2(+/-0.7) nm, respectively, showing that the SCR domains adopt a range of conformations including folded-back ones. The distance distribution function P(r) showed that the most commonly occurring distance in sCR1 is at 11.5 nm. Its maximum length of 55 nm is less than double those for CR2 or FH, even though sCR1 has twice the number of SCR domains compared to CR2 Sedimentation equilibrium experiments gave a mean molecular weight of 235 kDa for sCR1. This is consistent with the value of 245 kDa calculated from its composition including 14 N-linked oligosaccharide sites, and confirmed that sCR1 is a monomer in solution. Sedimentation velocity experiments gave a sedimentation coefficient of 5.8 S. From this, the frictional ratio (f/f(0)) of sCR1 was calculated to be 2.29, which is greater than those of 1.96 for CR2 and 1.77 for FH. The constrained scattering modelling of the sCR1 solution structure starting from homologous SCR domain structures generated 5000 trial conformationally randomised models, 43 of which gave good scattering fits to show that sCR1 has a partly folded-back structure. We conclude that the inter-SCR linkers show structural features in common with those in FH, but differ from those in CR2, and the SCR arrangement in CR1 will permit C3b or C4b to access all three ligand sites.


Subject(s)
Complement C3b/chemistry , Complement C4/chemistry , Peptide Fragments/chemistry , Protein Folding , Receptors, Complement/chemistry , Amino Acid Sequence , Binding Sites , Consensus Sequence , Conserved Sequence , Cysteine/chemistry , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Repetitive Sequences, Amino Acid , Rotation , Scattering, Radiation , Sequence Homology, Amino Acid , Tryptophan/chemistry , Ultracentrifugation , X-Rays
16.
J Med Primatol ; 36(6): 375-80, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17976043

ABSTRACT

BACKGROUND: Soluble complement receptor-1 (sCR1), a potent complement inhibitor, confers neuroprotection in a murine stroke model. Additional neuroprotective benefit is achieved by sLe x-glycosylation of sCR1. In an effort to translate sCR1-sLe x to clinical trials, we evaluated this agent in a primate stroke model. METHODS: Adult male baboons randomly received either sCR1-sLe x or vehicle. Stroke volume was assessed on day 3, and neurological examinations were conducted daily. Complement activity (CH50) was measured at 30 minute, 2, 6, 12 hour, 3, and 10 days post-ischemia. RESULTS: The experiment was terminated prematurely following an interim analysis. In a preliminary cohort (n = 3 per arm), infarct volume was greater in the treated animals. No difference in neurological score was found between groups. CH50 levels were significantly reduced in the sCR1sLe x-treated groups. A hypotensive response was also observed in animals treated with sCR1-sLe x. Conclusions Further work is necessary to explain the hypotensive response observed in primates prior to further clinical development of sCR1-sLe x.


Subject(s)
Disease Models, Animal , Neuroprotective Agents/administration & dosage , Papio anubis , Receptors, Complement/administration & dosage , Reperfusion Injury/prevention & control , Stroke/prevention & control , Animals , Brain Ischemia/prevention & control , Cerebral Infarction/prevention & control , Complement Hemolytic Activity Assay/veterinary , Drug Evaluation, Preclinical , Male , Random Allocation , Time Factors , Treatment Outcome
17.
Circulation ; 116(11 Suppl): I83-8, 2007 Sep 11.
Article in English | MEDLINE | ID: mdl-17846331

ABSTRACT

BACKGROUND: TP10, a potent inhibitor of complement activation during cardiopulmonary bypass (CPB) has been shown to significantly reduce the incidence of death and myocardial infarction (MI) in high-risk male patients undergoing cardiac surgery. However, the effect of TP10 in females was undefined because of the limited number of females studied. To examine the possibility of a gender effect, this phase 2 multi-center trial was undertaken to determine whether TP10 would also limit ischemic damage in a larger sample size of high-risk females undergoing cardiac surgery on cardiopulmonary bypass (CPB). METHODS AND RESULTS: This prospective, double-blind, placebo-controlled, multi-center trial involved 297 high-risk (urgent surgery, CABG + Valve, reoperations, ejection fraction <30%) female patients randomized to receive a 5 mg/kg dose of TP10 (n=150) or placebo (n=147) as a 30-minute intravenous infusion before surgery. The primary end point was the incidence of death or MI at 28 days after surgery. Complement activation was assessed by levels of CH50 and SC5b-9 during and after CPB. TP10 was well tolerated and there were no differences in the safety profiles of the 2 groups. Although TP10 effectively suppressed complement activation (at 2 hours after CPB CH50 (mean+SD % change from baseline) 50+/-17% placebo versus 4+/-14% TP10; P=0.0001; SC5b-9 (ng/mL) 917+/-1067 placebo versus 204+/-79 TP10; P=0.0001), there was no difference in the primary end point between the groups (17% placebo versus 21% TP10; P=0.2550). CONCLUSIONS: The benefits of TP10 appear to be gender-related. and mechanisms other than complement activation may be responsible for myocardial injury in high-risk female patients during cardiac surgery on CPB.


Subject(s)
Cardiac Surgical Procedures , Complement System Proteins/metabolism , Receptors, Complement/therapeutic use , Sex Characteristics , Aged , Cardiac Surgical Procedures/methods , Double-Blind Method , Female , Humans , Male , Middle Aged
18.
J Neurosurg ; 105(4): 595-601, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17044564

ABSTRACT

OBJECT: Postischemic cerebral inflammatory injury has been extensively investigated in an effort to develop effective neuroprotective agents. The complement cascade has emerged as an important contributor to postischemic neuronal injury. Soluble complement receptor Type 1 (sCR1), a potent inhibitor of complement activation, has been shown to reduce infarct volume and improve functional outcome after murine stroke. Given numerous high-profile failures to translate promising antiinflammatory strategies from the laboratory to the clinic and given the known species-specificity of the complement cascade, the authors sought to evaluate the neuroprotective effect of sCR1 in a nonhuman primate model of stroke. METHODS: A total of 48 adult male baboons (Papio anubis) were randomly assigned to receive 15 mg/kg of sCR1 or vehicle. The animals were subjected to 75 minutes of middle cerebral artery occlusion/reperfusion. Perioperative blood samples were analyzed for total complement activity by using a CH50 assay. Infarct volume and neurological scores were assessed at the time the animals were killed, and immunohistochemistry was used to determine cerebral drug penetration and C1q deposition. An interim futility analysis led to termination of the trial after study of 12 animals. Total serum complement activity was significantly depressed in the sCR1-treated animals compared with the controls. Immunostaining also demonstrated sCR1 deposition in the ischemic hemispheres of treated animals. Despite these findings, there were no significant differences in infarct volume or neurological score between the sCR1--and vehicle-treated cohorts. CONCLUSIONS: A preischemic bolus infusion of sCR1, the most effective means of administration in mice, was not neuroprotective in a primate model. This study illustrates the utility of a translational primate model of stroke in the assessment of promising antiischemic agents prior to implementation of large-scale clinical trials.


Subject(s)
Brain/blood supply , Disease Models, Animal , Infarction, Middle Cerebral Artery/immunology , Neuroprotective Agents/administration & dosage , Receptors, Complement 3b/administration & dosage , Reperfusion Injury/immunology , Animals , Brain/immunology , Brain/pathology , Complement C1q/analysis , Drug Evaluation, Preclinical , Immunoenzyme Techniques , Infarction, Middle Cerebral Artery/pathology , Male , Papio anubis , Reperfusion Injury/pathology
19.
Circulation ; 110(11 Suppl 1): II274-9, 2004 Sep 14.
Article in English | MEDLINE | ID: mdl-15364875

ABSTRACT

BACKGROUND: This study was undertaken to determine whether soluble human complement receptor type 1 (TP10), a potent inhibitor of complement activation, would reduce morbidity and mortality in high-risk patients undergoing cardiac surgery on cardiopulmonary bypass (CPB). METHODS: This was a randomized multicenter, prospective, placebo-controlled, double-blind study in which 564 high-risk patients undergoing cardiac surgery on CPB received an intravenous bolus of TP10 (1, 3, 5, 10 mg/kg) or placebo immediately before CPB. The primary endpoint was the composite events of death, myocardial infarction (MI), prolonged (> or =24 hours) intra-aortic balloon pump support (IABP), and prolonged intubation. RESULTS: TP10 significantly inhibited complement activity after 10 to 15 minutes of CPB and this inhibition persisted for 3 days postoperatively. However, there was no difference in the primary endpoint between the 2 groups (33.7% placebo versus 31.4% TP10; P=0.31). The primary composite endpoint was, however, reduced in all male TP10 patients by 30% (P=0.025). TP10 reduced the incidence of death or MI in males by 36% (P=0.026), the incidence of death or MI in CABG males by 43% (P=0.043) and the need for prolonged IABP support in male CABG and valve patients by 100% (P=0.019). There was, however, no improvement seen in female TP10 patients. There were no significant differences in adverse events between the groups. CONCLUSIONS: TP10 effectively inhibits complement activation during CPB; however, this was not associated with an improvement in the primary endpoint of the study. Nevertheless, TP10 did significantly decrease the incidence of mortality and MI in male patients.


Subject(s)
Cardiac Surgical Procedures , Cardiopulmonary Bypass/adverse effects , Complement Activation/drug effects , Myocardial Ischemia/prevention & control , Receptors, Complement/therapeutic use , Aged , Cardiac Surgical Procedures/mortality , Complement C3a/biosynthesis , Complement Membrane Attack Complex/biosynthesis , Double-Blind Method , Female , Humans , Infections/epidemiology , Injections, Intravenous , Intra-Aortic Balloon Pumping/statistics & numerical data , Length of Stay/statistics & numerical data , Male , Middle Aged , Myocardial Infarction/diagnosis , Myocardial Infarction/epidemiology , Myocardial Infarction/etiology , Myocardial Ischemia/etiology , Myocardial Ischemia/immunology , Prospective Studies , Receptors, Complement/administration & dosage , Sex Factors , Solubility , Treatment Outcome
20.
Glycobiology ; 14(10): 883-93, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15190008

ABSTRACT

Recombinant soluble human complement receptor type 1 (sCR1) is a highly glycosylated glycoprotein intended for use as a drug to treat ischemia-reperfusion injury and other complement-mediated diseases and injuries. sCR1-sLe(x) produced in the FT-VI-expressing mutant CHO cell line LEC11 exists as a heterogeneous mixture of glycoforms, a fraction of which include structures with one or more antennae terminated by the sialyl Lewis X (sLe(x)) [Neu5Acalpha2-3Galbeta1-4(Fucalpha1-3)GlcNAc]) epitope. Such multivalent presentation of sLe(x) was shown previously to effectively target sCR1 to activated endothelial cells expressing E-selectin. Here, we describe the use of the soluble, recombinant alpha2-3 sialyltransferase ST3Gal-III and the alpha1-3 fucosyltransferase FT-VI in vitro to introduce sLe(x) moieties onto the N-glycan chains of sCR1 overexpressed in standard CHO cell lines. The product (sCR1-S/F) of these in vitro enzymatic glycan remodeling reactions performed at the 10-g scale has approximately 14 N-glycan chains per sCR1 molecule, comprised of biantennary (90%), triantennary (8.5%), and tetraantennary (1.5%) structures, nearly all of whose antennae terminate with sLe(x) moieties. sCR1-S/F retained complement inhibitory activity and, in comparison with sCR1-sLe(x) produced in the LEC11 cell line, contained twice the number of sLe(x) moieties per mole glycoprotein, exhibited a twofold increase in area under the intravenous clearance curve in a rat pharmacokinetic model, and exhibited a 10-fold increase in affinity for E-selectin in an in vitro binding assay. These results demonstrate that in vitro glycosylation of the sCR1 drug product reduces heterogeneity of the glycan profile, improves pharmacokinetics, and enhances carbohydrate-mediated binding to E-selectin.


Subject(s)
Oligosaccharides/chemistry , Receptors, Complement/biosynthesis , Receptors, Complement/chemistry , Animals , CHO Cells , Cricetinae , E-Selectin/metabolism , Fucosyltransferases/metabolism , Fucosyltransferases/pharmacology , Glycosylation , Humans , Oligosaccharides/genetics , Oligosaccharides/metabolism , Polysaccharides/chemistry , Polysaccharides/genetics , Polysaccharides/metabolism , Rats , Receptors, Complement/genetics , Receptors, Complement/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sialyl Lewis X Antigen , Sialyltransferases/chemistry , Sialyltransferases/metabolism , Sialyltransferases/pharmacology , beta-Galactoside alpha-2,3-Sialyltransferase
SELECTION OF CITATIONS
SEARCH DETAIL
...