Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Pharm X ; 5: 100157, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36687375

ABSTRACT

Antibody-based T cell-activating biologics are promising therapeutic medicines being developed for a number of indications, mainly in the oncology field. Among those, T cell bispecific antibodies are designed to bind one tumor-specific antigen and the T cell receptor at the same time, leading to a robust T cell response against the tumor. Although their unique format and the versatility of the CrossMab technology allows for the generation of safer molecules in an efficient manner, product-related variants cannot be completely avoided. Therefore, it is of extreme importance that both a manufacturing process that limits or depletes product-related impurities, as well as a thorough analytical characterization are in place, starting from the development of the manufacturing cell line until the assessment of potential toxicities. Here, we describe such an end-to-end approach to minimize, quantify and control impurities and -upon their functional characterization- derive specifications that allow for the release of clinical material.

2.
J Exp Med ; 215(3): 859-876, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29436396

ABSTRACT

Depletion of immunosuppressive tumor-associated macrophages (TAMs) or reprogramming toward a proinflammatory activation state represent different strategies to therapeutically target this abundant myeloid population. In this study, we report that inhibition of colony-stimulating factor-1 receptor (CSF-1R) signaling sensitizes TAMs to profound and rapid reprogramming in the presence of a CD40 agonist before their depletion. Despite the short-lived nature of macrophage hyperactivation, combined CSF-1R+CD40 stimulation of macrophages is sufficient to create a proinflammatory tumor milieu that reinvigorates an effective T cell response in transplanted tumors that are either responsive or insensitive to immune checkpoint blockade. The central role of macrophages in regulating preexisting immunity is substantiated by depletion experiments, transcriptome analysis of ex vivo sorted TAMs, and gene expression profiling of whole tumor lysates at an early treatment time point. This approach enabled the identification of specific combination-induced changes among the pleiotropic activation spectrum of the CD40 agonist. In patients, CD40 expression on human TAMs was detected in mesothelioma and colorectal adenocarcinoma.


Subject(s)
Immunity , Macrophages/immunology , Neoplasms/immunology , Neoplasms/pathology , Animals , CD40 Antigens/agonists , CD40 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Inflammation/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , Phenotype , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/metabolism
3.
FEBS J ; 285(4): 763-776, 2018 02.
Article in English | MEDLINE | ID: mdl-28941174

ABSTRACT

Despite decades of research, cancer remains a devastating disease and new treatment options are needed. Today cancer is acknowledged as a multifactorial disease not only comprising of aberrant tumor cells but also the associated stroma including tumor vasculature, fibrotic plaques, and immune cells that interact in a complex heterotypic interplay. Myeloid cells represent one of the most abundant immune cell population within the tumor stroma and are equipped with a broad functional repertoire that promotes tumor growth by suppressing cytotoxic T cell activity, stimulating neoangiogenesis and tissue remodeling. Therefore, myeloid cells have become an attractive target for pharmacological intervention. In this review, we summarize the pharmacological approaches to therapeutically target tumor-associated myeloid cells with a focus on advanced programs that are clinically evaluated. In addition, for each therapeutic strategy, the preclinical rationale as well as advantages and challenges from a drug development perspective are discussed.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Development , Myeloid Cells/drug effects , Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Humans , Neoplasms/metabolism , Neoplasms/pathology
5.
Cancer Cell ; 30(3): 377-390, 2016 09 12.
Article in English | MEDLINE | ID: mdl-27622331

ABSTRACT

Effective cancer immunotherapy requires overcoming immunosuppressive tumor microenvironments. We found that local nitric oxide (NO) production by tumor-infiltrating myeloid cells is important for adoptively transferred CD8(+) cytotoxic T cells to destroy tumors. These myeloid cells are phenotypically similar to inducible nitric oxide synthase (NOS2)- and tumor necrosis factor (TNF)-producing dendritic cells (DC), or Tip-DCs. Depletion of immunosuppressive, colony stimulating factor 1 receptor (CSF-1R)-dependent arginase 1(+) myeloid cells enhanced NO-dependent tumor killing. Tumor elimination via NOS2 required the CD40-CD40L pathway. We also uncovered a strong correlation between survival of colorectal cancer patients and NOS2, CD40, and TNF expression in their tumors. Our results identify a network of pro-tumor factors that can be targeted to boost cancer immunotherapies.


Subject(s)
Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Neoplasms/immunology , Neoplasms/therapy , Nitric Oxide Synthase Type II/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Arginase/biosynthesis , Arginase/immunology , CD40 Antigens/immunology , CD40 Ligand/immunology , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/biosynthesis , Tumor Microenvironment , Tumor Necrosis Factor-alpha/biosynthesis
6.
PLoS One ; 11(7): e0159716, 2016.
Article in English | MEDLINE | ID: mdl-27463372

ABSTRACT

CD44, a transmembrane receptor reported to be involved in various cellular functions, is overexpressed in several cancer types and supposed to be involved in the initiation, progression and prognosis of these cancers. Since the sequence of events following the blockage of the CD44-HA interaction has not yet been studied in detail, we profiled xenograft tumors by RNA Sequencing to elucidate the mode of action of the anti-CD44 antibody RG7356. Analysis of tumor and host gene-expression profiles led us to the hypothesis that treatment with RG7356 antibody leads to an activation of the immune system. Using cytokine measurements we further show that this activation involves the secretion of chemo-attractants necessary for the recruitment of immune cells (i.e. macrophages) to the tumor site. We finally provide evidence for antibody-dependent cellular phagocytosis (ADCP) of the malignant cells by macrophages.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Hyaluronan Receptors/immunology , Macrophages/immunology , Animals , Cell Line, Tumor , Cytokines/genetics , Cytokines/metabolism , Humans , Mice , Mice, SCID , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Phagocytosis
7.
Oncotarget ; 7(22): 32532-42, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27081038

ABSTRACT

RG7356, a recombinant anti-CD44 immunoglobulin G1 humanized monoclonal antibody, inhibits cell adhesion and has been associated with macrophage activation in preclinical models. We report results of a phase I dose-escalation study of RG7356 in relapsed/refractory acute myeloid leukemia (AML).Eligible patients with refractory AML, relapsed AML after induction chemotherapy, or previously untreated AML not eligible for intensive chemotherapy were enrolled and received intravenous RG7356 at dosages ≤ 2400 mg every other week or ≤ 1200 mg weekly or twice weekly; dose escalation started at 300 mg.Forty-four patients (median age, 69 years) were enrolled. One dose-limiting toxicity occurred (grade 3 hemolysis exacerbation) after one 1200 mg dose (twice-weekly cohort). The majority of adverse events were mild/moderate. Infusion-related reactions occurred in 64% of patients mainly during cycle 1. Two patients experienced grade 3 drug-induced aseptic meningitis. Pharmacokinetics increased supraproportionally, suggesting a target-mediated drug disposition (TMDD) at ≥ 1200 mg. Two patients achieved complete response with incomplete platelet recovery or partial response, respectively. One patient had stable disease with hematologic improvement.RG7356 was generally safe and well tolerated. Maximum tolerated dose was not reached, but saturation of TMDD was achieved. The recommended dose for future AML evaluations is 2400 mg every other week.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Leukemia, Myeloid, Acute/drug therapy , Adult , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Young Adult
8.
Mol Cancer Ther ; 15(5): 946-57, 2016 05.
Article in English | MEDLINE | ID: mdl-27037412

ABSTRACT

Dysregulated cellular apoptosis and resistance to cell death are hallmarks of neoplastic initiation and disease progression. Therefore, the development of agents that overcome apoptosis dysregulation in tumor cells is an attractive therapeutic approach. Activation of the extrinsic apoptotic pathway is strongly dependent on death receptor (DR) hyperclustering on the cell surface. However, strategies to activate DR5 or DR4 through agonistic antibodies have had only limited clinical success. To pursue an alternative approach for tumor-targeted induction of apoptosis, we engineered a bispecific antibody (BsAb), which simultaneously targets fibroblast-activation protein (FAP) on cancer-associated fibroblasts in tumor stroma and DR5 on tumor cells. We hypothesized that bivalent binding to both FAP and DR5 leads to avidity-driven hyperclustering of DR5 and subsequently strong induction of apoptosis in tumor cells but not in normal cells. Here, we show that RG7386, an optimized FAP-DR5 BsAb, triggers potent tumor cell apoptosis in vitro and in vivo in preclinical tumor models with FAP-positive stroma. RG7386 antitumor efficacy was strictly FAP dependent, was independent of FcR cross-linking, and was superior to conventional DR5 antibodies. In combination with irinotecan or doxorubicin, FAP-DR5 treatment resulted in substantial tumor regression in patient-derived xenograft models. FAP-DR5 also demonstrated single-agent activity against FAP-expressing malignant cells, due to cross-binding of FAP and DR5 across tumor cells. Taken together, these data demonstrate that RG7386, a novel and potent antitumor agent in both mono- and combination therapies, overcomes limitations of previous DR5 antibodies and represents a promising approach to conquer tumor-associated resistance to apoptosis. Mol Cancer Ther; 15(5); 946-57. ©2016 AACR.


Subject(s)
Antibodies, Bispecific/metabolism , Antibodies, Bispecific/pharmacology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Gelatinases/metabolism , Membrane Proteins/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Serine Endopeptidases/metabolism , Animals , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antibody Affinity/immunology , Antineoplastic Agents/immunology , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Endopeptidases , Fibroblasts/drug effects , Fibroblasts/metabolism , Gelatinases/immunology , Humans , Membrane Proteins/immunology , Mice , Protein Binding/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Serine Endopeptidases/immunology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Proc Natl Acad Sci U S A ; 112(50): 15444-9, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26621726

ABSTRACT

Formative research suggests that a human embryonic stem cell-specific alternative splicing gene regulatory network, which is repressed by Muscleblind-like (MBNL) RNA binding proteins, is involved in cell reprogramming. In this study, RNA sequencing, splice isoform-specific quantitative RT-PCR, lentiviral transduction, and in vivo humanized mouse model studies demonstrated that malignant reprogramming of progenitors into self-renewing blast crisis chronic myeloid leukemia stem cells (BC LSCs) was partially driven by decreased MBNL3. Lentiviral knockdown of MBNL3 resulted in reversion to an embryonic alternative splice isoform program typified by overexpression of CD44 transcript variant 3, containing variant exons 8-10, and BC LSC proliferation. Although isoform-specific lentiviral CD44v3 overexpression enhanced chronic phase chronic myeloid leukemia (CML) progenitor replating capacity, lentiviral shRNA knockdown abrogated these effects. Combined treatment with a humanized pan-CD44 monoclonal antibody and a breakpoint cluster region - ABL proto-oncogene 1, nonreceptor tyrosine kinase (BCR-ABL1) antagonist inhibited LSC maintenance in a niche-dependent manner. In summary, MBNL3 down-regulation-related reversion to an embryonic alternative splicing program, typified by CD44v3 overexpression, represents a previously unidentified mechanism governing malignant progenitor reprogramming in malignant microenvironments and provides a pivotal opportunity for selective BC LSC detection and therapeutic elimination.


Subject(s)
Alternative Splicing/genetics , Cell Self Renewal/genetics , Human Embryonic Stem Cells/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Adult , Animals , Apoptosis/genetics , Blast Crisis/genetics , Blast Crisis/pathology , Bone Marrow/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Proliferation , Cell Survival , Cellular Reprogramming/genetics , Female , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Hematopoiesis , Humans , Hyaluronan Receptors/metabolism , Ligands , Male , Mice , Middle Aged , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pluripotent Stem Cells/cytology , Proto-Oncogene Mas
10.
Clin Cancer Res ; 21(12): 2753-62, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25762343

ABSTRACT

PURPOSE: CD44, a cell surface glycoprotein, plays important roles in the development, progression, and metastasis of various tumor types. The aim of this study was to investigate how the expression of CD44 isoforms influences the interaction with hyaluronic acid (HA) and how differential isoform expression impacts antitumoral responses in vivo to treatment with RG7356, a humanized anti-CD44 antibody inhibiting CD44-HA interaction. EXPERIMENTAL DESIGN: CD44 isoform expression on various tumor cell lines was analyzed by RNASeq while data on patients with different tumor types were obtained from the publicly available TCGA RNASeq dataset as well as a phase I clinical study (NCT01358903). We analyzed the link between HA production and CD44 isoform expression as well as the consequences of blocking the CD44-mediated cell adhesion to HA using RG7356. The correlation between CD44 isoform expression and antitumor response to RG7356 treatment was investigated in the corresponding murine xenograft in vivo models as well as in a subset of patients treated with RG7356 from a recently completed phase I clinical trial. RESULTS: CD44 isoform expression, in particular expression of CD44s, is associated with HA production and predicts response to treatment with RG7356 in tumor xenograft models. Furthermore, patient data suggest that CD44 isoform status is a potential predictive biomarker for clinical response to treatment with RG7356. CONCLUSIONS: We provide new insights into the close interplay between CD44 and HA and a potential biomarker to enrich patient responses to RG7356 in the clinic.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Hyaluronan Receptors/metabolism , Neoplasms/metabolism , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/therapeutic use , Disease Models, Animal , Female , Gene Expression , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Hyaluronan Receptors/genetics , Hyaluronic Acid/metabolism , Male , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Protein Binding , Protein Interaction Maps , Protein Isoforms , Transcription, Genetic , Transfection , Treatment Outcome , Xenograft Model Antitumor Assays
11.
Cancer Cell ; 25(6): 846-59, 2014 Jun 16.
Article in English | MEDLINE | ID: mdl-24898549

ABSTRACT

Macrophage infiltration has been identified as an independent poor prognostic factor in several cancer types. The major survival factor for these macrophages is macrophage colony-stimulating factor 1 (CSF-1). We generated a monoclonal antibody (RG7155) that inhibits CSF-1 receptor (CSF-1R) activation. In vitro RG7155 treatment results in cell death of CSF-1-differentiated macrophages. In animal models, CSF-1R inhibition strongly reduces F4/80(+) tumor-associated macrophages accompanied by an increase of the CD8(+)/CD4(+) T cell ratio. Administration of RG7155 to patients led to striking reductions of CSF-1R(+)CD163(+) macrophages in tumor tissues, which translated into clinical objective responses in diffuse-type giant cell tumor (Dt-GCT) patients.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Colonic Neoplasms/therapy , Macrophages/drug effects , Macrophages/immunology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/immunology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Cell Differentiation/physiology , Cell Line, Tumor , Clinical Trials, Phase I as Topic , Cohort Studies , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Female , Humans , Macaca fascicularis , Macrophages/cytology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Models, Molecular , Receptor, Macrophage Colony-Stimulating Factor/metabolism
12.
Mol Immunol ; 56(4): 488-96, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23911405

ABSTRACT

Ficolins activate the lectin pathway of the complement system upon binding to carbohydrate patterns on pathogens. To characterize the producer cells of ficolin-B the expression of mouse ficolin-B, the orthologue of human M-ficolin, was studied in macrophages and dendritic cells during differentiation from bone marrow cells, in primary granulocytes, and during differentiation of granulocytes derived from ER-Hoxb8 cells. Expression of ficolin-B mRNA declined in all myeloid cell types to low levels during terminal differentiation. However, in contrast to macrophages and dendritic cells, ficolin-B expression was enhanced upon activation in granulocytes. High expression of ficolin-B was observed in primary immature neutrophilic CD11b(+) Ly-6C(int) Ly-6G(high) granulocytes when isolated from the bone marrow, in particular during sepsis. Ficolin-B was demonstrated in lysates of primary granulocytes, ER-Hoxb8-derived granulocytes, bone marrow-derived macrophages, and dendritic cells. Native ficolin-B from cell lysates and supernatants of granulocytes activated the lectin pathway as measured by binding to MASP-2 and inducing C4 deposition. Specific staining demonstrated intra-cellular or cell associated ficolin-B protein in activated immature granulocytes deposited in a granular fashion. This study shows that ficolin-B is stored in and set free from immature granulocytic myeloid cells indicating a role in the early infection-induced cellular response of these inflammatory cells.


Subject(s)
Granulocytes/metabolism , Lectins/metabolism , Myeloid Cells/metabolism , Neutrophils/metabolism , Animals , Blotting, Western , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Line , Complement Activation , Dendritic Cells/cytology , Dendritic Cells/metabolism , Estrogens/pharmacology , Gene Expression/drug effects , Granulocytes/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lectins/genetics , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/metabolism , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Myeloid Cells/cytology , Neutrophils/cytology , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Ficolins
13.
Immunobiology ; 217(10): 982-5, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22459270

ABSTRACT

Ficolins and mannan-binding lectin recognize pathogen-associated molecular patterns and initiate the lectin pathway of complement activation via the associated serine proteases. In contrast to human ficolins and mouse ficolin-A, mouse ficolin-B has been considered incapable of complement activation. Dose-dependent binding of recombinant ficolin-B to immobilized GlcNAc, acetylated BSA, acetylated LDL, and fetuin was detected with ficolin-B-specific monoclonal antibodies. Recombinant ficolin-B bound to immobilized acetylated bovine serum albumin interacted with recombinant human mannan-binding lectin-associated serine protease-2, which led to C4 cleavage, thus demonstrating the capability of ficolin-B to activate the lectin pathway. Ficolin-B-specific monoclonal antibodies identified natural ficolin-B protein in lysates of mouse granulocytes isolated from the bone marrow. These results identify mouse ficolin-B as a functional member of the ficolin family activating complement via the lectin pathway.


Subject(s)
Lectins/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Animals , Cell Line , Complement Activation/immunology , Complement C4/immunology , Complement C4/metabolism , Complement System Proteins/immunology , Humans , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Mice , Mice, Inbred C57BL , Protein Binding/immunology , Recombinant Proteins/metabolism , Ficolins
14.
Immunobiology ; 213(3-4): 297-306, 2008.
Article in English | MEDLINE | ID: mdl-18406375

ABSTRACT

Ficolins are members of the collectin family of proteins which are able to recognize pathogen-associated molecular pattern (PAMP) on microbial surfaces. Upon binding to their specific PAMP, ficolins may trigger activation of the immune system by either binding to cellular receptors for collectins or by initiating activation of complement via the lectin pathway. For the latter, the human ficolins (i.e. L-, H- and M-ficolin) and murine ficolin-A were shown to associate with the lectin pathway-specific serine protease MBL-associated serine protease-2 (MASP-2) and catalyse its activation which in turn activates C4 and C4b-bound C2 to generate the C3 convertase C4b2a. There is mounting evidence underlining the lectin nature of ficolins with a wide range of carbohydrate moieties recognized on microbial surfaces. However, not all members of the ficolin family appear to act as lectin pathway recognition components. For example, murine ficolin-B does not associate with MASP-2 and appears to be absent in plasma and other humoral fluids. Its stringent cellular localization points to other functions within the immune response, possibly acting as an intracellular scavenger to target and facilitate clearance of PAMP-bearing debris. When comparing ficolin orthologues from different species, it appears evident that human, murine, and porcine ficolins differ in many aspects, a specific point that we aim to address in this review.


Subject(s)
Complement System Proteins/metabolism , Immune System , Immunity, Innate , Lectins/metabolism , Animals , Humans , Lectins/genetics , Lectins/physiology , Macrophages/microbiology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Mice , Models, Biological , Polymorphism, Genetic , Species Specificity , Swine , Ficolins
15.
J Endotoxin Res ; 12(2): 120-6, 2006.
Article in English | MEDLINE | ID: mdl-16690015

ABSTRACT

Ficolins are pattern-recognition molecules of the innate immune system able to trigger the lectin pathway of the complement activation upon binding to microbial surfaces. In humans, two plasma ficolins have been identified and characterized, whereas a third cell-associated ficolin (M-ficolin) was found on monocyte surfaces. The mouse homologue of M-ficolin is called ficolin B. Although the spatial-temporal expression patterns of mouse ficolins have been described recently, the subcellular localization of ficolin B protein is so far unknown. By using ficolin B-specific antibodies and confocal microscopy, we show that ficolin B is expressed within mouse peritoneal exudate macrophages and is co-localized with Lamp-1, a marker for lysosomes and late endosomes. In addition, the data indicate that ficolin B expression is up-regulated upon macrophage activation.


Subject(s)
Lectins/metabolism , Lysosomes/metabolism , Macrophages/metabolism , Animals , Blotting, Western , Bone Marrow/metabolism , Chinchilla , Female , Immunoprecipitation , Lectins/biosynthesis , Lectins/isolation & purification , Lysosomal Membrane Proteins/metabolism , Macrophage Activation , Macrophages/ultrastructure , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Spleen/metabolism , Ficolins
SELECTION OF CITATIONS
SEARCH DETAIL
...