Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Immunol ; 207(9): 2245-2254, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34561227

ABSTRACT

Targeting interactions between α4ß7 integrin and endothelial adhesion molecule MAdCAM-1 to inhibit lymphocyte migration to the gastrointestinal tract is an effective therapy in inflammatory bowel disease (IBD). Following lymphocyte entry into the mucosa, a subset of these cells expresses αEß7 integrin, which is expressed on proinflammatory lymphocytes, to increase cell retention. The factors governing lymphocyte migration into the intestinal mucosa and αE integrin expression in healthy subjects and IBD patients remain incompletely understood. We evaluated changes in factors involved in lymphocyte migration and differentiation within tissues. Both ileal and colonic tissue from active IBD patients showed upregulation of ICAM-1, VCAM-1, and MAdCAM-1 at the gene and protein levels compared with healthy subjects and/or inactive IBD patients. ß1 and ß7 integrin expression on circulating lymphocytes was similar across groups. TGF-ß1 treatment induced expression of αE on both ß7+ and ß7- T cells, suggesting that cells entering the mucosa independently of MAdCAM-1/α4ß7 can become αEß7+ ITGAE gene polymorphisms did not alter protein induction following TGF-ß1 stimulation. Increased phospho-SMAD3, which is directly downstream of TGF-ß, and increased TGF-ß-responsive gene expression were observed in the colonic mucosa of IBD patients. Finally, in vitro stimulation experiments showed that baseline ß7 expression had little effect on cytokine, chemokine, transcription factor, and effector molecule gene expression in αE+ and αE- T cells. These findings suggest cell migration to the gut mucosa may be altered in IBD and α4ß7-, and α4ß7+ T cells may upregulate αEß7 in response to TGF-ß once within the gut mucosa.


Subject(s)
Antigens, CD/metabolism , Inflammatory Bowel Diseases/immunology , Integrin alpha Chains/metabolism , Integrin beta Chains/metabolism , Intestinal Mucosa/immunology , Receptors, Lymphocyte Homing/metabolism , T-Lymphocytes/immunology , Adult , Aged , Cell Movement , Female , Humans , Integrin beta Chains/genetics , Male , Middle Aged , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism
2.
Cytometry A ; 91(1): 39-47, 2017 01.
Article in English | MEDLINE | ID: mdl-27632576

ABSTRACT

Mass cytometry is capable of measuring more than 40 distinct proteins on individual cells making it a promising technology for innovating biomarker discovery. However, in order for this potential to be fully realized, best practices in panel design need to be further defined in order to achieve consistency and reproducibility in data analysis. Of particular importance are controls that reveal, and panel design principles that mitigate the effects of signal interference or overlap. We observed a disparity between the staining profiles of two noncompeting anti- integrin ß7 mAbs and hypothesized that signal interference was responsible. A mass-minus-one (MMO) control was applied and demonstrated that signal overlap caused the perceived interclonal discrepancy in ß7 expression. Panel redesign in consideration of mass-cytometry specific interference dynamics dramatically improved concordance between both mAbs by redistributing background signals caused by overlap. These studies visualize how signal overlap can complicate mass cytometry data interpretation and demonstrate how the rational distribution of interference can greatly improve panel design and data quality. © 2016 International Society for Advancement of Cytometry.


Subject(s)
Antibodies, Monoclonal/immunology , Flow Cytometry/methods , Integrin beta Chains/biosynthesis , Leukocytes, Mononuclear/metabolism , Antibodies, Monoclonal/chemistry , Gene Expression Regulation , Humans , Integrin beta Chains/immunology , Leukocytes, Mononuclear/ultrastructure
3.
Inflamm Bowel Dis ; 28(9): 1348-1356, 2022 09 01.
Article in English | MEDLINE | ID: mdl-34849918

ABSTRACT

BACKGROUND: Etrolizumab, a humanized anti-ß7 antibody, has not been studied in children. Here, we evaluate the pharmacokinetics, pharmacodynamics, and safety of etrolizumab in children with inflammatory bowel disease. METHODS: Patients age 4 to 17 years with moderately to severely active ulcerative colitis or Crohn's disease were randomized 1:1 to receive 1.5mg/kg of etrolizumab subcutaneously every 4 weeks (q4w) or 3.0mg/kg every 8 weeks (q8w) for 16 weeks in this open-label phase 1 trial. Pharmacokinetics, pharmacodynamics, safety, and efficacy were assessed. RESULTS: Of the 24 patients treated, 21 completed the study. In the groups of 1.5mg/kg q4w and 3.0mg/kg q8w, respectively, mean (SD) maximum concentration (Cmax) was 9.8 (4.86) µg/mL and 18.1 (6.25) µg/mL; and mean (SD) area under the curve within a dosing interval (AUCtau) was 167 (86.9) and 521 (306) µg·day/mL after the last dose. The Cmax increased dose proportionally. The AUC over an 8-week period was slightly higher in the 3.0mg/kg q8w dose group. Median half-life was similar for both dosing regimens. Median numbers of free ß7high gut-homing T and B cell subsets declined below 10% of baseline, confirming ß7 target engagement and complete/near-complete receptor occupancy. Adverse events were consistent with the safety profile in adults. Approximately 60% of patients achieved a clinical response. CONCLUSIONS: Etrolizumab showed a dose-proportional increase in Cmax and a slightly greater than dose-proportional increase in AUCtau. Both regimens achieved complete/near-complete ß7 receptor occupancy, with a similar relationship to concentration as adults. Etrolizumab was well tolerated and demonstrated clinical activity in children.


Subject(s)
Colitis, Ulcerative , Crohn Disease , Inflammatory Bowel Diseases , Adolescent , Adult , Antibodies, Monoclonal, Humanized , Child , Child, Preschool , Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Humans , Inflammatory Bowel Diseases/drug therapy
4.
Clin Cancer Res ; 14(9): 2775-84, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451245

ABSTRACT

PURPOSE: We generated a humanized antibody, HuLuc63, which specifically targets CS1 (CCND3 subset 1, CRACC, and SLAMF7), a cell surface glycoprotein not previously associated with multiple myeloma. To explore the therapeutic potential of HuLuc63 in multiple myeloma, we examined in detail the expression profile of CS1, the binding properties of HuLuc63 to normal and malignant cells, and the antimyeloma activity of HuLuc63 in preclinical models. EXPERIMENTAL DESIGN: CS1 was analyzed by gene expression profiling and immunohistochemistry of multiple myeloma samples and numerous normal tissues. HuLuc63-mediated antimyeloma activity was tested in vitro in antibody-dependent cellular cytotoxicity (ADCC) assays and in vivo using the human OPM2 xenograft model in mice. RESULTS: CS1 mRNA was expressed in >90% of 532 multiple myeloma cases, regardless of cytogenetic abnormalities. Anti-CS1 antibody staining of tissues showed strong staining of myeloma cells in all plasmacytomas and bone marrow biopsies. Flow cytometric analysis of patient samples using HuLuc63 showed specific staining of CD138+ myeloma cells, natural killer (NK), NK-like T cells, and CD8+ T cells, with no binding detected on hematopoietic CD34+ stem cells. HuLuc63 exhibited significant in vitro ADCC using primary myeloma cells as targets and both allogeneic and autologous NK cells as effectors. HuLuc63 exerted significant in vivo antitumor activity, which depended on efficient Fc-CD16 interaction as well as the presence of NK cells in the mice. CONCLUSIONS: These results suggest that HuLuc63 eliminates myeloma cells, at least in part, via NK-mediated ADCC and shows the therapeutic potential of targeting CS1 with HuLuc63 for the treatment of multiple myeloma.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lymphocyte Subsets/metabolism , Multiple Myeloma/drug therapy , Plasma Cells/metabolism , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Female , Gene Expression Profiling , Humans , Killer Cells, Natural/immunology , Lymphocyte Subsets/cytology , Mice , Mice, SCID , Multiple Myeloma/immunology , Plasma Cells/cytology , Receptors, Immunologic/genetics , Signaling Lymphocytic Activation Molecule Family , Xenograft Model Antitumor Assays
5.
Front Pharmacol ; 10: 39, 2019.
Article in English | MEDLINE | ID: mdl-30774593

ABSTRACT

Background: Anti-integrin therapy is a new frontline strategy in the treatment of inflammatory bowel diseases (IBD). The anti-ß7 integrin antibody etrolizumab is currently being investigated for safety and efficacy in Crohn's disease (CD) and ulcerative colitis (UC) in several phase III trials. Mechanistically, etrolizumab is known to block ß7 integrin ligand binding and reduces intestinal trafficking of ß7-expressing cells. Etrolizumab blocks ß7 integrin ligand binding and reduces ß7-positive lymphocyte migration and retention in the inflamed gut mucosa, but the exact mechanisms by which this inhibition occurs are not fully understood. Methods: Cellular effects of etrolizumab or etrolizumab surrogate antibody (etrolizumab-s) were investigated in cell culture models and analyzed by flow cytometry, fluorescence microscopy, ImageStream®, stimulated emission depletion (STED) microscopy and functional dynamic in vitro adhesion assays. Moreover, effects on α4ß7 integrin were compared with the pharmacodynamically similar antibody vedolizumab. Results: As demonstrated by several different approaches, etrolizumab and etrolizumab-s treatment led to internalization of ß7 integrin. This resulted in impaired dynamic adhesion to MAdCAM-1. Internalized ß7 integrin localized in endosomes and re-expression of ß7 was dependent on de novo protein synthesis. In vitro etrolizumab treatment did not lead to cellular activation or cytokine secretion and did not induce cytotoxicity. Internalization of α4ß7 integrin was increased with etrolizumab compared with vedolizumab. Discussion: Our data suggest that etrolizumab does not elicit secondary effector functions on the single cell level. Integrin internalization may be an important mechanism of action of etrolizumab, which might explain some but not all immunological effects observed with etrolizumab.

6.
Br J Pharmacol ; 176(19): 3805-3818, 2019 10.
Article in English | MEDLINE | ID: mdl-31270798

ABSTRACT

BACKGROUND AND PURPOSE: Polatuzumab vedotin is an antibody-drug conjugate (ADC) being developed for non-Hodgkin's lymphoma. It contains a humanized anti-CD79b IgG1 monoclonal antibody linked to monomethyl auristatin E (MMAE), an anti-mitotic agent. Polatuzumab vedotin binds to human CD79b only. Therefore, a surrogate ADC that binds to cynomolgus monkey CD79b was used to determine CD79b-mediated pharmacological effects in the monkey and to enable first-in-human clinical trials. EXPERIMENTAL APPROACH: Polatuzumab vedotin, the surrogate ADC, and the corresponding antibodies were evaluated in different assays in vitro and in animals. In vitro assessments included binding to peripheral blood mononuclear cells from different species, binding to a human and monkey CD79b-expressing cell line, binding to human Fcγ receptors, and stability in plasma across species. In vivo, ADCs were assessed for anti-tumour activity in mice, pharmacokinetics/pharmacodynamics in monkeys, and toxicity in rats and monkeys. KEY RESULTS: Polatuzumab vedotin and surrogate ADC bind with similar affinity to human and cynomolgus monkey B cells, respectively. Comparable in vitro plasma stability, in vivo anti-tumour activity, and mouse pharmacokinetics were also observed between the surrogate ADC and polatuzumab vedotin. In monkeys, only the surrogate ADC showed B-cell depletion and B-cell-mediated drug disposition, but both ADCs showed similar MMAE-driven myelotoxicity, as expected. CONCLUSIONS AND IMPLICATIONS: The suitability of the surrogate ADC for evaluation of CD79b-dependent pharmacology was demonstrated, and anti-tumour activity, pharmacokinetics/pharmacodynamics, and toxicity data with both ADCs supported the entry of polatuzumab vedotin into clinical trials.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Burkitt Lymphoma/drug therapy , CD79 Antigens/antagonists & inhibitors , Immunoconjugates/pharmacology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Binding Sites/drug effects , Burkitt Lymphoma/pathology , CD79 Antigens/immunology , Cell Line , Dose-Response Relationship, Drug , Female , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Macaca fascicularis , Male , Mice , Mice, SCID , Molecular Conformation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Rats , Rats, Sprague-Dawley , Receptors, IgG , Structure-Activity Relationship
7.
J Clin Pharmacol ; 58(3): 386-398, 2018 03.
Article in English | MEDLINE | ID: mdl-29178491

ABSTRACT

Etrolizumab, a humanized monoclonal antibody, specifically binds to the ß7 subunit of the heterodimeric integrins α4ß7 and αEß7. Pharmacokinetic (PK) and pharmacodynamic (PD) data were collected from an etrolizumab phase 1 trial in patients with moderate to severe ulcerative colitis (UC). We developed a mechanism-based model to simultaneously describe the kinetics of serum etrolizumab concentration and free ß7 receptors on circulating intestinal-homing CD4+ T lymphocytes. Included in the analysis were 38 phase 1 UC patients who received single or 3 monthly doses of etrolizumab intravenously or subcutaneously across a dose range of 0.3 to 10 mg/kg. A quasi-steady-state target-mediated drug disposition model was developed to describe the dynamic interaction between serum etrolizumab concentration and free ß7 receptors on intestinal-homing CD4+ T lymphocytes in UC patients. The time profiles of serum etrolizumab and absolute counts of ß7+ lymphocytes (expressed as percentage of baseline level) were well described by the quasi-steady-state target-mediated drug disposition model. The model was able to characterize the maximum drug occupancy of ß7 receptors on intestinal-homing CD4+ T lymphocytes and the concentration-dependent duration of occupancy. The 90% effective concentration for etrolizumab to saturate the ß7 receptors on intestinal homing CD4+ T cells was 1.3 µg/mL. PK and PD profiles predicted by the model were consistent with observations from a subsequent phase 2 study. In conclusion, an integrated PK/PD model developed in this analysis reasonably described serum etrolizumab PK profiles and the relationship between PK and PD (free ß7 receptors on circulating intestinal-homing CD4+ T lymphocytes) in UC patients.


Subject(s)
Antibodies, Monoclonal, Humanized/blood , Antibodies, Monoclonal, Humanized/pharmacology , Colitis, Ulcerative/blood , Colitis, Ulcerative/drug therapy , Integrins/blood , Models, Biological , Antibodies, Monoclonal, Humanized/pharmacokinetics , Cohort Studies , Colitis, Ulcerative/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Doublecortin Domain Proteins , Female , Gastrointestinal Agents/blood , Gastrointestinal Agents/pharmacokinetics , Gastrointestinal Agents/pharmacology , Humans , Immunologic Factors/blood , Immunologic Factors/pharmacokinetics , Immunologic Factors/pharmacology , Male , Microtubule-Associated Proteins , Neuropeptides
9.
Br J Pharmacol ; 174(8): 628-640, 2017 04.
Article in English | MEDLINE | ID: mdl-28009435

ABSTRACT

BACKGROUND AND PURPOSE: CD22 and CD79b are cell-surface receptors expressed on B-cell-derived malignancies such as non-Hodgkin's lymphoma (NHL). An anti-mitotic agent, monomethyl auristatin E, was conjugated to anti-CD22 and anti-CD79b antibodies to develop target-specific therapies for NHL. The mechanism of action (MOA) and pharmacological and pharmacokinetic (PK) profiles of these antibody-drug conjugates (ADCs) were investigated in cynomolgus monkeys. EXPERIMENTAL APPROACH: Animals were administered anti-CD22 or anti-CD79b ADCs, respective unconjugated antibodies or vehicle. Pharmacodynamic effects on total and proliferating B cells and serum PK were then assessed. Antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) of the ADCs were evaluated in vitro. KEY RESULTS: Depletion of B cells was observed after administration of either ADC or the respective unconjugated antibodies. An extended duration of depletion was observed in animals administered ADCs. Similarly, preferential depletion of proliferating B cells in blood and germinal centre B cells in spleen were only observed in animals administered ADCs. Serum PK profiles of ADCs and respective unconjugated antibodies were comparable. In vitro, anti-human CD22 and anti-human CD79b antibodies showed no or only moderate ADCC activity, respectively; neither antibody had CDC activity. CONCLUSIONS AND IMPLICATIONS: The findings support the proposed MOA: initial depletion of total B cells by antibody-mediated opsonization, followed by preferential, sustained depletion of proliferating B cells by the auristatin conjugate due to its anti-mitotic action. Delivering potent anti-mitotic agents to B cells via the specificity of monoclonal antibodies provides a means to eliminate pathogenic B cells in NHL with improved risk-benefit profiles over traditional chemotherapeutics.


Subject(s)
Antibodies/immunology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , CD79 Antigens/immunology , Oligopeptides/pharmacology , Sialic Acid Binding Ig-like Lectin 2/immunology , Animals , Antigen-Antibody Reactions , B-Lymphocytes/immunology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Macaca fascicularis , Male , Structure-Activity Relationship
10.
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
11.
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
SELECTION OF CITATIONS
SEARCH DETAIL