Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Mol Cancer Ther ; 21(8): 1326-1336, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35666803

ABSTRACT

Probody therapeutics (Pb-Txs) are conditionally activated antibody-drug conjugates (ADCs) designed to remain inactive until proteolytically activated in the tumor microenvironment, enabling safer targeting of antigens expressed in both tumor and normal tissue. Previous attempts to target CD71, a highly expressed tumor antigen, have failed to establish an acceptable therapeutic window due to widespread normal tissue expression. This study evaluated whether a probody-drug conjugate targeting CD71 can demonstrate a favorable efficacy and tolerability profile in preclinical studies for the treatment of cancer. CX-2029, a Pb-Tx conjugated to maleimido-caproyl-valine-citrulline-p-aminobenzyloxycarbonyl-monomethyl auristatin E, was developed as a novel cancer therapeutic targeting CD71. Preclinical studies were performed to evaluate the efficacy and safety of this anti-CD71 PDC in patient-derived xenograft (PDX) mouse models and cynomolgus monkeys, respectively. CD71 expression was detected at high levels by IHC across a broad range of tumor and normal tissues. In vitro, the masked Pb-Tx form of the anti-CD71 PDC displayed a >50-fold reduced affinity for binding to CD71 on cells compared with protease-activated, unmasked anti-CD71 PDC. Potent in vivo tumor growth inhibition (stasis or regression) was observed in >80% of PDX models (28/34) at 3 or 6 mg/kg. Anti-CD71 PDC remained mostly masked (>80%) in circulation throughout dosing in cynomolgus monkeys at 2, 6, and 12 mg/kg and displayed a 10-fold improvement in tolerability compared with an anti-CD71 ADC, which was lethal. Preclinically, anti-CD71 PDC exhibits a highly efficacious and acceptable safety profile that demonstrates the utility of the Pb-Tx platform to target CD71, an otherwise undruggable target. These data support further clinical development of the anti-CD71 PDC CX-2029 as a novel cancer therapeutic.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Lead , Macaca fascicularis/metabolism , Mice , Neoplasms/drug therapy , Tumor Microenvironment , Xenograft Model Antitumor Assays
2.
Blood ; 114(13): 2721-9, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19633198

ABSTRACT

Here we describe the generation of an antibody-drug conjugate (ADC) consisting of a humanized anti-CD79b antibody that is conjugated to monomethylauristatin E (MMAE) through engineered cysteines (THIOMABs) by a protease cleavable linker. By using flow cytometry, we detected the surface expression of CD79b in almost all non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia patients, suggesting that anti-CD79b-vcMMAE could be widely used in these malignancies. By using NHL cell lines to simulate a patient population we discovered that a minimal cell-surface expression level of CD79b was required for in vitro activity. Within the subpopulation of cell lines above this minimal threshold, we found that sensitivity to free MMAE, mutation of cancer genes, and cell doubling time were poorly correlated with in vitro activity; however, the expression level of BCL-XL was correlated with reduced sensitivity to anti-CD79b-vcMMAE. This observation was supported by in vivo data showing that a Bcl-2 family inhibitor, ABT-263, strikingly enhanced the activity of anti-CD79b-vcMMAE. Furthermore, anti-CD79b-vcMMAE was significantly more effective than a standard-of-care regimen, R-CHOP (ie, rituximab with a single intravenous injection of 30 mg/kg cyclophosphamide, 2.475 mg/kg doxorubicin, 0.375 mg/kg vincristine, and oral dosing of 0.15 mg/kg prednisone once a day for 5 days), in 3 xenograft models of NHL. Together, these data suggest that anti-CD79b-vcMMAE could be broadly efficacious for the treatment of NHL.


Subject(s)
CD79 Antigens/immunology , Lymphoma, Non-Hodgkin/drug therapy , Oligopeptides/therapeutic use , Animals , Antibodies, Anti-Idiotypic/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/drug effects , Female , Humans , Immunoconjugates/therapeutic use , Lymphoma, Non-Hodgkin/pathology , Mice , Mice, Inbred ICR , Mice, SCID , Oligopeptides/chemistry , Treatment Outcome , Tumor Burden , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
PLoS One ; 9(10): e109366, 2014.
Article in English | MEDLINE | ID: mdl-25285661

ABSTRACT

Inhibiting NAD biosynthesis by blocking the function of nicotinamide phosphoribosyl transferase (NAMPT) is an attractive therapeutic strategy for targeting tumor metabolism. However, the development of drug resistance commonly limits the efficacy of cancer therapeutics. This study identifies mutations in NAMPT that confer resistance to a novel NAMPT inhibitor, GNE-618, in cell culture and in vivo, thus demonstrating that the cytotoxicity of GNE-618 is on target. We determine the crystal structures of six NAMPT mutants in the apo form and in complex with various inhibitors and use cellular, biochemical and structural data to elucidate two resistance mechanisms. One is the surprising finding of allosteric modulation by mutation of residue Ser165, resulting in unwinding of an α-helix that binds the NAMPT substrate 5-phosphoribosyl-1-pyrophosphate (PRPP). The other mechanism is orthosteric blocking of inhibitor binding by mutations of Gly217. Furthermore, by evaluating a panel of diverse small molecule inhibitors, we unravel inhibitor structure activity relationships on the mutant enzymes. These results provide valuable insights into the design of next generation NAMPT inhibitors that offer improved therapeutic potential by evading certain mechanisms of resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Cytokines/antagonists & inhibitors , Cytokines/chemistry , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/chemistry , Catalytic Domain , Cell Line, Tumor , Cytokines/genetics , Humans , Models, Molecular , Mutation , Nicotinamide Phosphoribosyltransferase/genetics
4.
Clin Cancer Res ; 19(24): 6912-23, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24097869

ABSTRACT

PURPOSE: We sought to identify predictive biomarkers for a novel nicotinamide phosphoribosyltransferase (NAMPT) inhibitor. EXPERIMENTAL DESIGN: We use a NAMPT inhibitor, GNE-617, to evaluate nicotinic acid rescue status in a panel of more than 400 cancer cell lines. Using correlative analysis and RNA interference (RNAi), we identify a specific biomarker for nicotinic acid rescue status. We next determine the mechanism of regulation of expression of the biomarker. Finally, we develop immunohistochemical (IHC) and DNA methylation assays and evaluate cancer tissue for prevalence of the biomarker across indications. RESULTS: Nicotinate phosphoribosyltransferase (NAPRT1) is necessary for nicotinic acid rescue and its expression is the major determinant of rescue status. We demonstrate that NAPRT1 promoter methylation accounts for NAPRT1 deficiency in cancer cells, and NAPRT1 methylation is predictive of rescue status in cancer cell lines. Bisulfite next-generation sequencing mapping of the NAPRT1 promoter identified tumor-specific sites of NAPRT1 DNA methylation and enabled the development of a quantitative methylation-specific PCR (QMSP) assay suitable for use on archival formalin-fixed paraffin-embedded tumor tissue. CONCLUSIONS: Tumor-specific promoter hypermethylation of NAPRT1 inactivates one of two NAD salvage pathways, resulting in synthetic lethality with the coadministration of a NAMPT inhibitor. NAPRT1 expression is lost due to promoter hypermethylation in most cancer types evaluated at frequencies ranging from 5% to 65%. NAPRT1-specific immunohistochemical or DNA methylation assays can be used on archival formalin paraffin-embedded cancer tissue to identify patients likely to benefit from coadministration of a Nampt inhibitor and nicotinic acid.


Subject(s)
Cytokines/metabolism , Heterocyclic Compounds, 2-Ring/administration & dosage , Neoplasms/genetics , Niacin/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Pentosyltransferases/metabolism , Sulfones/administration & dosage , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cytokines/antagonists & inhibitors , Cytokines/genetics , DNA Methylation/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/pathology , Niacin/administration & dosage , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/genetics , Pentosyltransferases/antagonists & inhibitors , Pentosyltransferases/deficiency , Promoter Regions, Genetic
5.
Neoplasia ; 15(10): 1151-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24204194

ABSTRACT

Nicotinamide adenine dinucleotide (NAD) is a critical metabolite that is required for a range of cellular reactions. A key enzyme in the NAD salvage pathway is nicotinamide phosphoribosyl transferase (NAMPT), and here, we describe GNE-618, an NAMPT inhibitor that depletes NAD and induces cell death in vitro and in vivo. While cells proficient for nicotinic acid phosphoribosyl transferase (NAPRT1) can be protected from NAMPT inhibition as they convert nicotinic acid (NA) to NAD independent of the salvage pathway, this protection only occurs if NA is added before NAD depletion. We also demonstrate that tumor cells are unable to generate NAD by de novo synthesis as they lack expression of key enzymes in this pathway, thus providing a mechanistic rationale for the reliance of tumor cells on the NAD salvage pathway. Identifying tumors that are sensitive to NAMPT inhibition is one potential way to enhance the therapeutic effectiveness of an NAMPT inhibitor, and here, we show that NAMPT, but not NAPRT1, mRNA and protein levels inversely correlate with sensitivity to GNE-618 across a panel of 53 non-small cell lung carcinoma cell lines. Finally, we demonstrate that GNE-618 reduced tumor growth in a patient-derived model, which is thought to more closely represent heterogeneous primary patient tumors. Thus, we show that dependence of tumor cells on the NAD salvage pathway renders them sensitive to GNE-618 in vitro and in vivo, and our data support further evaluation of the use of NAMPT mRNA and protein levels as predictors of overall sensitivity.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , NAD/metabolism , Pentosyltransferases/antagonists & inhibitors , Pyrazoles/pharmacology , Sulfones/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Death/drug effects , Cell Line, Tumor , Heterografts , Humans , Lung Neoplasms/drug therapy , Mice, Nude , Pentosyltransferases/genetics , Pyrazoles/therapeutic use , Sulfones/therapeutic use
6.
Mol Cancer Ther ; 12(7): 1255-65, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23598530

ABSTRACT

Antibody-drug conjugates (ADC), potent cytotoxic drugs linked to antibodies via chemical linkers, allow specific targeting of drugs to neoplastic cells. We have used this technology to develop the ADC DCDT2980S that targets CD22, an antigen with expression limited to B cells and the vast majority of non-Hodgkin lymphomas (NHL). DCDT2980S consists of a humanized anti-CD22 monoclonal IgG1 antibody with a potent microtubule-disrupting agent, monomethyl auristatin E (MMAE), linked to the reduced cysteines of the antibody via a protease cleavable linker, maleimidocaproyl-valine-citrulline-p-aminobenzoyloxycarbonyl (MC-vc-PAB). We describe the efficacy, safety, and pharmacokinetics of DCDT2980S in animal models to assess its potential as a therapeutic for the treatment of B-cell malignancies. We did not find a strong correlation between in vitro or in vivo efficacy and CD22 surface expression, nor a correlation of sensitivity to free drug and in vitro potency. We show that DCDT2980S was capable of inducing complete tumor regression in xenograft mouse models of NHL and can be more effective than rituximab plus combination chemotherapy at drug exposures that were well tolerated in cynomolgus monkeys. These results suggest that DCDT2980S has an efficacy, safety, and pharmacokinetics profile that support potential treatment of NHL.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Immunoconjugates/pharmacology , Lymphoma, B-Cell/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Oligopeptides/pharmacology , Sialic Acid Binding Ig-like Lectin 2/immunology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Female , Humans , Immunoconjugates/pharmacokinetics , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Macaca fascicularis , Mice , Mice, Inbred ICR , Mice, SCID , Random Allocation , Xenograft Model Antitumor Assays
7.
Mol Cancer Ther ; 11(10): 2222-32, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22807577

ABSTRACT

Fc receptor-like 5 (FcRL5/FcRH5/IRTA2/CD307) is a surface protein expressed selectively on B cells and plasma cells. We found that FcRL5 was expressed at elevated levels on the surface of plasma cells from the bone marrow of patients diagnosed with multiple myeloma. This prevalence in multiple myeloma and narrow pattern of normal expression indicate that FcRL5 could be a target for antibody-based therapies for multiple myeloma, particularly antibody-drug conjugates (ADC), potent cytotoxic drugs linked to antibodies via specialized chemical linkers, where limited expression on normal tissues is a key component to their safety. We found that FcRL5 is internalized upon antibody binding, indicating that ADCs to FcRL5 could be effective. Indeed, we found that FcRL5 ADCs were efficacious in vitro and in vivo but the unconjugated antibody was not. The two most effective consisted of our anti-FcRL5 antibody conjugated through cysteines to monomethylauristatin E (MMAE) by a maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl (MC-vcPAB) linker (anti-FcRL5-MC-vcPAB-MMAE) or conjugated via lysines to the maytansinoid DM4 through a disulfide linker (anti-FcRL5-SPDB-DM4). These two ADCs were highly effective in vivo in combination with bortezomib or lenalidomide, drugs in use for the treatment of multiple myeloma. These data show that the FcRL5 ADCs described herein show promise as an effective treatment for multiple myeloma.


Subject(s)
Antibodies, Neoplasm/immunology , Antineoplastic Agents/therapeutic use , Immunoconjugates/therapeutic use , Molecular Targeted Therapy , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Receptors, Cell Surface/immunology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Endocytosis/drug effects , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Mice , Mice, SCID , Receptors, Fc , Reproducibility of Results , Treatment Outcome
8.
Sci Transl Med ; 3(74): 74ra22, 2011 Mar 16.
Article in English | MEDLINE | ID: mdl-21411738

ABSTRACT

The primary function of B cells, critical components of the adaptive immune response, is to produce antibodies against foreign antigens, as well as to perform isotype class switching, which changes the heavy chain of an antibody so that it can interact with different repertoires of effector cells. CD40 is a member of the tumor necrosis factor superfamily of cell surface receptors that transmits survival signals to B cells. In contrast, in B cell cancers, stimulation of CD40 signaling results in a heterogeneous response in which cells can sometimes undergo cell death in response to treatment, depending on the system studied. We found an association between sensitivity to CD40 stimulation and mutation of the tumor suppressor p53 in a panel of non-Hodgkin's lymphoma cell lines. Consistent with p53's tumor suppressor role, we found that higher levels of intrinsic DNA damage and increased proliferation rates, as well as higher levels of BCL6, a transcriptional repressor proto-oncogene, were associated with sensitivity to CD40 stimulation. In addition, CD40 treatment-resistant cell lines were sensitized to CD40 stimulation after the introduction of DNA-damaging agents. Using gene expression analysis, we also showed that resistant cell lines exhibited a preexisting activated CD40 pathway and that an mRNA expression signature comprising CD40 target genes predicted sensitivity and resistance to CD40-activating agents in cell lines and mouse xenograft models. Finally, the gene signature predicted tumor shrinkage and progression-free survival in patients with diffuse large B cell lymphoma treated with dacetuzumab, a monoclonal antibody with partial CD40 agonist activity. These data show that CD40 pathway activation status may be useful in predicting the antitumor activity of CD40-stimulating therapeutic drugs.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD40 Antigens/immunology , Immunotherapy/methods , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Animals , Antibodies, Monoclonal, Humanized , B-Lymphocytes/immunology , CD40 Antigens/genetics , CD40 Ligand/genetics , CD40 Ligand/immunology , Cell Line, Tumor , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Mice , Microarray Analysis , Proto-Oncogene Mas , Transplantation, Heterologous , Tumor Suppressor Protein p53/immunology
9.
Mol Cancer Ther ; 8(10): 2937-46, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19808977

ABSTRACT

Antibodies directed against B cells are in use for the treatment of non-Hodgkin's lymphoma and autoimmune disorders. The B-cell-restricted surface antigen CD79b, a signaling component of the B-cell receptor, has been shown as a promising antibody target in mouse efficacy models of systemic lupus erythematosus. Anti-CD79b antibody-drug conjugates (ADC), cytotoxic drugs linked through specialized chemical linkers to antibodies, are effective in mouse xenograft models of non-Hodgkin's lymphoma. We were interested in evaluating the systemic effects of anti-CD79b antibodies and ADCs in normal animals as a step toward the development of these molecules as therapeutics. As we were unable to identify any cell surface binding anti-human CD79b antibodies that were cross-reactive to other species, we developed an antibody to cynomolgus monkey (Macaca fascicularis) CD79b (anti-cyCD79b). The anti-cynomolgus antibody, anti-cyCD79b (10D10), and the maytansine (tubulin inhibitor)-conjugated ADC, anti-cyCD79b (10D10)-MCC-DM1, were administered to cynomolgus monkeys at approximately 30 mg/kg (6,000 microg DM1/m(2)) for two doses 3 weeks apart. Anti-cyCD79b and anti-cyCD79b-MCC-DM1 resulted in peripheral blood B-cell depletion of approximately 65% and approximately 94%, respectively. In addition, anti-cyCD79b-MCC-DM1 resulted in near-complete absence of splenic germinal centers, an observation supporting an effect on dividing B cells. Both molecules were well tolerated, with minimal findings for the antibody and findings for the ADC limited to the lymphoid and hematopoietic systems, liver, and peripheral nerves. These preclinical data suggest that targeting CD79b with antibodies or ADCs may provide safe and effective therapies for B-cell malignancies and autoimmune diseases.


Subject(s)
Antibodies/immunology , Antineoplastic Agents/pharmacology , CD79 Antigens/immunology , Amino Acid Sequence , Animals , Antibody Formation/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CD79 Antigens/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Cross Reactions/drug effects , Flow Cytometry , Humans , Immune Tolerance/drug effects , Macaca fascicularis/blood , Macaca fascicularis/immunology , Maytansine/pharmacology , Mice , Molecular Sequence Data , Spleen/drug effects , Spleen/immunology , Spleen/pathology , Xenograft Model Antitumor Assays
10.
Cancer Res ; 69(6): 2358-64, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19258515

ABSTRACT

Antibody-drug conjugates (ADC), potent cytotoxic drugs covalently linked to antibodies via chemical linkers, provide a means to increase the effectiveness of chemotherapy by targeting the drug to neoplastic cells while reducing side effects. Here, we systematically examine the potential targets and linker-drug combinations that could provide an optimal ADC for the treatment for non-Hodgkin's lymphoma. We identified seven antigens (CD19, CD20, CD21, CD22, CD72, CD79b, and CD180) for potential treatment of non-Hodgkin's lymphoma with ADCs. ADCs with cleavable linkers mediated in vivo efficacy via all these targets; ADCs with uncleavable linkers were only effective when targeted to CD22 and CD79b. In target-independent safety studies in rats, the uncleavable linker ADCs showed reduced toxicity, presumably due to the reduced release of free drug or other toxic metabolites into the circulation. Thus, our data suggest that ADCs with cleavable linkers work on a broad range of targets, and for specific targets, ADCs with uncleavable linkers provide a promising opportunity to improve the therapeutic window for ADCs in humans.


Subject(s)
Antineoplastic Agents/administration & dosage , Immunotoxins/pharmacology , Lymphoma, Non-Hodgkin/drug therapy , Maytansine/analogs & derivatives , Oligopeptides/administration & dosage , Sulfhydryl Compounds/administration & dosage , Animals , Antigens, CD/biosynthesis , Antigens, CD/immunology , Antineoplastic Agents/pharmacokinetics , B-Lymphocytes/immunology , Cross-Linking Reagents/administration & dosage , Cross-Linking Reagents/pharmacokinetics , Female , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Lymphoma, Non-Hodgkin/immunology , Maytansine/administration & dosage , Maytansine/pharmacokinetics , Mice , Mice, Inbred ICR , Mice, SCID , Oligopeptides/pharmacokinetics , Rats , Sulfhydryl Compounds/pharmacokinetics , Xenograft Model Antitumor Assays
11.
Blood ; 110(2): 616-23, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17374736

ABSTRACT

Targeting cytotoxic drugs to cancer cells using antibody-drug conjugates (ADCs), particularly those with stable linkers between the drug and the antibody, could be an effective cancer treatment with low toxicity. However, for stable-linker ADCs to be effective, they must be internalized and degraded, limiting potential targets to surface antigens that are trafficked to lysosomes. CD79a and CD79b comprise the hetrodimeric signaling component of the B-cell receptor, and are attractive targets for the use of ADCs because they are B-cell-specific, expressed in non-Hodgkin lymphomas (NHL), and are trafficked to a lysosomal-like compartment as part of antigen presentation. We show here that the stable-linker ADCs anti-CD79b-MCC-DM1 and anti-CD79b-MC-MMAF are capable of target-dependent killing of nonHodgkin lymphoma cell lines in vitro. Further, these 2 ADCs are equally effective as low doses in xenograft models of follicular, mantle cell, and Burkitt lymphomas, even though several of these cell lines express relatively low levels of CD79b in vivo. In addition, we demonstrate that anti-CD79b ADCs were more effective than anti-CD79a ADCs and that, as hypothesized, anti-CD79b antibodies downregulated surface B-cell receptor and were trafficked to the lysosomal-like major histocompatibility complex class II-positive compartment MIIC. These results suggest that anti-CD79b-MCC-DM1 and anti-CD79b-MC-MMAF are promising therapeutics for the treatment of NHL.


Subject(s)
Antibodies/therapeutic use , CD79 Antigens/immunology , Immunoconjugates/therapeutic use , Lymphoma, Non-Hodgkin/immunology , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Female , Flow Cytometry , HLA-D Antigens/immunology , Humans , Lysosomes/immunology , Mice , Mice, SCID , Receptors, Antigen, B-Cell/immunology , Transplantation, Heterologous
12.
Int Immunol ; 18(9): 1363-73, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16849395

ABSTRACT

A new family of Ig domain receptors referred to as the immune receptor translocation-associated (IRTA) proteins, FcR homologs (FcRHs) or FcR-like that are expressed in lymphoid cells has been recently described. RNA expression analysis suggests that FcRH1-5/IRTA1-5 are expressed exclusively in subsets of the B-cell compartment. We generated mAbs to FcRH1-5/IRTA1-5 and examined their protein expression pattern in normal tissue and in chronic lymphocytic leukemia (CLL) cells. Our data indicated that FcRH1-5/IRTA1-5 were expressed in B-cell sub-populations; however, in some cases, the protein was not expressed in the same B-cell populations as suggested by the RNA expression analysis. FcRH1/IRTA5 was expressed throughout the B-cell lineage starting at the pro-B-cell stage but was down-regulated in plasma cells. FcRH2/IRTA4 was expressed preferentially in memory B cells. FcRH3/IRTA3 was expressed at low levels in naive, germinal center (GC) and memory B cells but was also expressed in NK cells. FcRH4/IRTA1 was expressed in a sub-population of memory B cells associated with mucosal tissue. FcRH5/IRTA2 was expressed in mature B cells and memory B cells and down-regulated in GC cells and, unlike all other B-cell-specific markers, maintained its expression in plasma cells from tonsil, spleen and bone marrow. We examined the expression of FcRH1-5/IRTA1-5 on the surface of CLL cells and found a similar pattern of expression on CLL cells as in the normal mature B cells, except for FcRH3/IRTA3 which was up-regulated in CLL.


Subject(s)
B-Lymphocytes/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Receptors, Cell Surface/biosynthesis , Receptors, Immunologic/biosynthesis , Animals , Flow Cytometry , Humans , Immunohistochemistry , Killer Cells, Natural/metabolism , Mice , Polymerase Chain Reaction , Receptors, Fc
SELECTION OF CITATIONS
SEARCH DETAIL