Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Pharmacol Res ; 178: 106147, 2022 04.
Article in English | MEDLINE | ID: mdl-35227891

ABSTRACT

CTLA4-Ig is a potent costimulatory blocker that inhibits T cell activation during alloimmune inflammation and increases graft survival and function. CTLA4-Ig-mediated immunosuppression has been demonstrated to support transplant function in various clinical trials and preclinical settings, but its effects on the balance between regulatory T cells (Tregs) and effector T cells (Teffs), as well as complement activation, are less well investigated. In the present study, we proposed to investigate the effects of CTLA4-Ig mediated immunosuppression on the phase of immunotolerance and the subsequent graft microvascular and epithelial repair during the progression of subepithelial fibrosis in a mouse model of orthotopic trachea transplantation. Briefly, CTLA4-Ig treated allografts (2 mg/kg, I.P.), untreated allografts, and syngrafts were serially monitored for peripheral FOXP3+ Tregs, antibody-mediated complement activation (C3d and C4d), tissue oxygenation, donor-recipient microvascular blood flow, and subsequent tissue remodeling following transplantation. Our data demonstrate that CTLA4-Ig mediated immunosuppression significantly results in late increases in both peripheral CD4+/CD8+ FOXP3+ Tregs and serum IL-10, but prevents the microvascular deposition of IgG, complement factor C3d, and epithelial C4d respectively, which proportionally improved blood flow and tissue oxygenation in the graft and, thus, promotes graft repair. Also, it restored the airway lumen, epithelium, and prevented the progression of subepithelial collagen deposition up to 90 days after transplantation. This study demonstrates that CTLA4-Ig-mediated immunosuppression potentially modulates both effector response and a late surge of regulatory activity to preserve graft microvasculature and rescue allograft from sustained hypoxia and ischemia and thereby limits subepithelial fibrosis.


Subject(s)
CTLA-4 Antigen , Graft Rejection , Graft Survival , Abatacept/pharmacology , Abatacept/therapeutic use , Animals , CTLA-4 Antigen/administration & dosage , CTLA-4 Antigen/immunology , Fibrosis , Forkhead Transcription Factors , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival/immunology , Immunosuppression Therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , Trachea/transplantation
2.
Mol Cancer Ther ; 19(2): 513-524, 2020 02.
Article in English | MEDLINE | ID: mdl-31871267

ABSTRACT

Cytotoxic T-lymphocyte antigen 4 (CTLA4)-FasL, a homo-hexameric signal converter protein, is capable of inducing robust apoptosis in malignant cells of the B-cell lineage expressing its cognate B7 and Fas targets, while sparing nonmalignant ones. This fusion protein's striking proapoptotic efficacy stems from its complementary abilities to coordinately activate apoptotic signals and abrogate antiapoptotic ones. A limiting factor in translating FasL or Fas receptor agonists into the clinic has been lethal hepatotoxicity. Here, we establish CTLA4-FasL's in vivo efficacy in multiple murine and xenograft models, for both systemic and subcutaneous tumors. Significantly, good laboratory practice (GLP) toxicology studies in mice indicate that CTLA4-FasL given repeatedly at doses up to five times the effective dose was well-tolerated and resulted in no significant adverse events. An equivalent single dose of CTLA4-FasL administered to nonhuman primates was also well-tolerated, albeit with a moderate dose-dependent leukopenia that was completely reversible. Interestingly, monkey peripheral blood mononuclear cells were more sensitive to CTLA4-FasL-induced apoptosis when tested in vitro. In both species, there was short-term elevation in serum levels of IL6, IL2, and IFNγ, although this was not associated with clinical signs of proinflammatory cytokine release, and further, this cytokine elevation could be completely prevented by dexamethasone premedication. Liver toxicity was not observed in either species, as confirmed by serum liver enzyme levels and histopathologic assessment. In conclusion, CTLA4-FasL emerges from animal model studies as an effective and safe agent for targeted FasL-mediated treatment of B7-expressing aggressive B-cell lymphomas.


Subject(s)
CTLA-4 Antigen/administration & dosage , Fas Ligand Protein/administration & dosage , Recombinant Fusion Proteins/pharmacology , Amino Acid Sequence , Animals , CTLA-4 Antigen/immunology , Fas Ligand Protein/adverse effects , Fas Ligand Protein/immunology , Fas Ligand Protein/pharmacokinetics , Female , Humans , Jurkat Cells , Macaca fascicularis , Mice , Mice, Inbred BALB C , Mice, Nude , Primates , Random Allocation , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics , Xenograft Model Antitumor Assays
3.
Actas dermo-sifiliogr. (Ed. impr.) ; 110(1): 43-49, ene.-feb. 2019. ilus, tab
Article in Spanish | IBECS | ID: ibc-176886

ABSTRACT

Las reacciones granulomatosas "sarcoidosis-like" secundarias a ipilimumab afectarán simultáneamente a múltiples órganos y característicamente se presentarán semanas o meses después de haber iniciado el tratamiento. Paciente varón de 67 años, quien consulta por reacción cutánea granulomatosa a nivel de tronco, extremidades y rostro. Esta se presentó 18 meses después de haber iniciado tratamiento con ipilimumab, medicamento pautado por el diagnóstico de un melanoma metastásico. Reportamos reacción granulomatosa de presentación exclusivamente cutánea, como efecto adverso medicamentoso tardío secundario al tratamiento con ipilimumab. Así mismo resaltamos la necesidad de realizar en los pacientes tratados con ipilimumab un seguimiento prolongado


Granulomatous sarcoidosis-like reactions affecting multiple organ systems at one time have infrequently been described within weeks to months after initiation of Ipilimumab. We present the first case of a 67-year-old man with isolated cutaneous granulomatous reaction involving the trunk, extremities, and face after eighteen months of treatment with ipilimumab for metastatic melanoma. This case documents the eruption of isolated cutaneous granulomatous reaction as a late treatment-related adverse effect of ipilimumab, highlighting the importance of adequate, prolonged follow-up


Subject(s)
Humans , Male , Female , Middle Aged , Aged , Melanoma/complications , Melanoma/diagnosis , Immunotherapy/methods , Neoplasm Metastasis/diagnosis , Antibodies, Monoclonal/administration & dosage , Sarcoidosis/diagnosis , Granuloma/diagnosis , CTLA-4 Antigen/administration & dosage , Pruritus/diagnosis , Radiography, Thoracic , Granuloma/pathology , Adrenal Cortex Hormones/administration & dosage
4.
Annu Rev Med ; 70: 409-424, 2019 01 27.
Article in English | MEDLINE | ID: mdl-30379596

ABSTRACT

The driver and passenger mutations accumulated in the process of malignant transformation offer an adequate spectrum of immune visible alterations to the cellular proteome and resulting peptidome to render these cancers targetable-and, in theory, rejectable-by the host T cell immune response. In addition, cancers often overexpress tissue-specific and developmental antigens to which immune tolerance is incomplete. Sometimes, virally transferred oncogenes drive malignant transformation and remain expressed throughout the cancer. Despite this state of antigenic sufficiency, cancer grows progressively and overcomes all efforts of the host immune system to contain it. While therapeutic cancer vaccination can mobilize high frequencies of tumor-specific T cells, these responses remain subject to intratumoral attenuation. Antibody modulation of T cell function through checkpoint blockade or costimulatory activation can restore survival, proliferation, and effector function to these tumor-infiltrating T cells and convert otherwise subtherapeutic vaccines into potentially curative cancer immunotherapeutics.


Subject(s)
CTLA-4 Antigen/administration & dosage , Cancer Vaccines/administration & dosage , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Animals , CTLA-4 Antigen/immunology , Cancer Vaccines/immunology , Cell Transformation, Neoplastic/immunology , Female , Forecasting , Humans , Immunologic Factors/administration & dosage , Male , Neoplasms/pathology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Risk Assessment , T-Lymphocytes/immunology , Treatment Outcome
5.
Clin Drug Investig ; 38(10): 967-976, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30143953

ABSTRACT

BACKGROUND AND OBJECTIVE: Immuno-oncology therapies represent a new treatment opportunity for patients affected by metastatic melanoma. The purpose of this study was to estimate the costs of immune-related adverse events (irAEs) associated with the new anti-PD1 immuno-oncology therapies, with the anti-CTLA-4 immuno-oncology therapy and with the combined therapy (CTLA4 + anti-PD1) in patients affected by metastatic melanoma. MATERIALS AND METHODS: A probabilistic cost-of-illness (COI) model was developed to estimate the management costs of grade ≥ 3 adverse events associated with the new anti-PD1 therapies (pembrolizumab and nivolumab), the anti-CTLA-4 therapy (ipilimumab) and the combined therapy CTLA4 + anti-PD1 (nivolumab + ipilimumab) for the treatment of patients with metastatic melanoma from the National Health Service (NHS) perspective in Italy. Identification of the epidemiological and cost parameters was carried out through a systematic literature review (SLR). Univariate and probabilistic sensitivity analyses were performed to account for uncertainty and variation in the model results. RESULTS: The model estimated a cost associated with the management of grade ≥ 3 immune-related adverse events in patients with metastatic melanoma equal to €176.2 (95% CI 63.5-335.0) for anti-CTLA-4 therapy, €48.6 (95% CI 40.1-58.5) for the new anti-PDI therapies and €276.8 (95% CI 240.4-316.2) for the combined therapy. Among the innovative therapies for the considered metastatic melanoma, the combined therapy was the most expensive innovative treatment in terms of event management of immune-related grade ≥ 3 adverse events. CONCLUSION: This study may represent a useful tool to understand the economic burden associated with the management of irAEs associated with patients affected by metastatic melanoma.


Subject(s)
Antineoplastic Agents, Immunological/economics , Costs and Cost Analysis/methods , Drug-Related Side Effects and Adverse Reactions/economics , Melanoma/economics , Therapies, Investigational/economics , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/economics , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , CTLA-4 Antigen/administration & dosage , Drug-Related Side Effects and Adverse Reactions/epidemiology , Humans , Ipilimumab/administration & dosage , Ipilimumab/adverse effects , Ipilimumab/economics , Italy/epidemiology , Melanoma/drug therapy , Melanoma/epidemiology , Nivolumab/administration & dosage , Nivolumab/adverse effects , Nivolumab/economics , Therapies, Investigational/adverse effects
6.
Oncol Rep ; 39(2): 511-518, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29207143

ABSTRACT

Cytotoxic T­lymphocyte antigen­4 (CTLA­4) is a critical negative regulator of immune responses. CTLA­4 is rapidly upregulated following T­cell activation, and then binds to B7 molecules with a higher affinity than CD28. CTLA­4 may abolish the initiation of the responses of T cells by raising the threshold of signals required for full activation of T cells, and it also may terminate ongoing T-cell responses. This regulatory role has led to the development of monoclonal antibodies (mAbs) designed to block CTLA­4 activity for enhancing immune responses against cancer. mAbs have several disadvantages including high production cost and unstable behavior. Nanobodies (Nbs) are single­domain antigen­binding fragments derived from the camelid heavy­chain antibodies, which are highly attractive in cancer immunotherapy due to their small size, high specificity, and stability. We selected CTLA­4­specific Nbs from a high quality dromedary camel immune library by phage display technology. Four positive colonies were sequenced and classified based on the amino acids sequences in the CDR3 region. These Nbs recognized unique epitopes on CTLA­4 and displayed high binding rates when used on PHA­stimulated human T cells. Treatment of B16 melanoma­bearing C57BL/6 mice with anti­CTLA­4 nanobody 16 (Nb16) delayed melanoma growth and prolonged the survival time of mice. These data indicate that anti­CTLA­4 Nbs selected from a high quality phage display library may be effective for the treatment of patients with tumors.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Cancer Vaccines/administration & dosage , Cell Surface Display Techniques/methods , Melanoma, Experimental/drug therapy , Single-Domain Antibodies/administration & dosage , Animals , CTLA-4 Antigen/administration & dosage , CTLA-4 Antigen/chemistry , Camelus , Cancer Vaccines/metabolism , Cancer Vaccines/pharmacology , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Immunization , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/pharmacology , Treatment Outcome , Xenograft Model Antitumor Assays
7.
Clin Pharmacol Drug Dev ; 5(5): 408-25, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27410490

ABSTRACT

ASP2408 is a next-generation anti-cytotoxic T lymphocyte antigen-4 fusion protein engineered for improved CD86 binding affinity as a treatment for rheumatoid arthritis (RA). In 72 healthy subjects (n = 6/treatment), ASP2408 was administered as single ascending doses intravenously at 0.003 to 10.0 mg/kg or subcutaneously at 0.3 to 3.0 mg/kg. It showed decreased clearance and prolonged half-life with increasing doses, consistent with target-mediated disposition. The apparent bioavailability was 36.3%-56.7% across single subcutaneous doses. Sixteen RA patients (n = 8/treatment) on stable methotrexate received 3 × 3.0 mg/kg subcutaneously every 4 weeks or every 2 weeks. Similar to single-dose treatment, ASP2408 concentrations peaked 2 to 3 days postdose, with a median t1/2 of approximately 8 days. Using CD86 receptor occupancy (RO) as a mechanistic biomarker, ASP2408 demonstrated dose-dependent binding to its target. ASP2408 3.0 mg/kg subcutaneously every 4 weeks and every 2 weeks led to a mean %CD86 RO ≥ 74.7% and ≥ 81.5%, respectively, within each dosing interval. ASP2408 was well tolerated across studies with no evidence of dose-limiting toxicity or clinically significant changes in clinical laboratory test results, vital signs, or 12-lead electrocardiograms. ASP2408 elicited antidrug antibodies in the majority of patients, but with no clinical sequelae.


Subject(s)
Antirheumatic Agents/administration & dosage , Arthritis, Rheumatoid/drug therapy , CTLA-4 Antigen/administration & dosage , Immunoconjugates/administration & dosage , Immunoglobulin G/administration & dosage , T-Lymphocytes/immunology , Administration, Intravenous , Adult , Aged , Antibodies/immunology , Antirheumatic Agents/adverse effects , Antirheumatic Agents/pharmacokinetics , B7-2 Antigen/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Female , Half-Life , Humans , Immunoconjugates/adverse effects , Immunoconjugates/pharmacokinetics , Immunoglobulin G/adverse effects , Injections, Subcutaneous , Male , Methotrexate/administration & dosage , Middle Aged , Young Adult
8.
EBioMedicine ; 6: 50-58, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27211548

ABSTRACT

Tumor indoleamine 2,3-dioxygenase (IDO) promotes immunosuppression by direct action on effector T cells and Tregs and through recruitment, expansion and activation of myeloid-derived suppressor cells (MDSCs). Targeting of MDSCs is clinically being explored as a therapeutic strategy, though optimal targeting strategies and biomarkers predictive of response are presently unknown. Maturation and tumor recruitment of MDSCs are dependent on signaling through the receptor tyrosine kinase CSF-1R on myeloid cells. Here, we show that MDSCs are the critical cell population in IDO-expressing B16 tumors in mediating accelerated tumor outgrowth and resistance to immunotherapy. Using a clinically relevant drug, we show that inhibition of CSF-1R signaling can functionally block tumor-infiltrating MDSCs and enhance anti-tumor T cell responses. Furthermore, inhibition of CSF-1R sensitizes IDO-expressing tumors to immunotherapy with T cell checkpoint blockade, and combination of CSF-1R blockade with IDO inhibitors potently elicits tumor regression. These findings provide evidence for a critical and functional role for MDSCs on the in vivo outcome of IDO-expressing tumors.


Subject(s)
Colonic Neoplasms/drug therapy , Immunotherapy/methods , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Melanoma, Experimental/drug therapy , Myeloid-Derived Suppressor Cells/drug effects , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , CTLA-4 Antigen/administration & dosage , Cell Line, Tumor , Colonic Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Melanoma, Experimental/metabolism , Mice , Molecular Targeted Therapy , Programmed Cell Death 1 Receptor/administration & dosage , Xenograft Model Antitumor Assays
9.
Immunopharmacol Immunotoxicol ; 37(6): 520-6, 2015.
Article in English | MEDLINE | ID: mdl-26555302

ABSTRACT

Adenoviruses (Ad) are once potential and promising vectors for gene delivery, but the immunogenicity attenuates its transfer efficiency. Cytotoxic T lymphocyte antigen 4 (CTLA-4) can inhibit T cell immunity. Thus, we aimed to study the effect of CTLA-4 in the process of Ad-mediated gene transfer. The C57BL/6 mice were injected by Ad vectors at twice, and CTLA-4 was administrated after the first Ad injection. Then, the CD3(+)CD4(+) T cells and circulating levels of IL-2, IL-4, and anti-Ad IgG were decreased by CTLA-4, while Ad generated immune responses. The green fluorescence protein (GFP) expressions of tissues were enhanced by CTLA-4 till injection of Ad at twice. Our results indicate that CTLA-4 can inhibit humoral and cellular immunity by adenovirus generation to enhance GFP delivery, and provide a potential way to assist in Ad-mediated gene transfer.


Subject(s)
Adenoviridae/immunology , CTLA-4 Antigen/administration & dosage , Genetic Vectors/administration & dosage , Green Fluorescent Proteins/administration & dosage , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Animals , Female , Gene Targeting/methods , Gene Transfer Techniques , Green Fluorescent Proteins/biosynthesis , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Mice , Mice, Inbred C57BL
11.
Cell ; 158(5): 989-999, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25131989

ABSTRACT

Latent reservoirs of HIV-1-infected cells are refractory to antiretroviral therapies (ART) and remain the major barrier to curing HIV-1. Because latently infected cells are long-lived, immunologically invisible, and may undergo homeostatic proliferation, a "shock and kill" approach has been proposed to eradicate this reservoir by combining ART with inducers of viral transcription. However, all attempts to alter the HIV-1 reservoir in vivo have failed to date. Using humanized mice, we show that broadly neutralizing antibodies (bNAbs) can interfere with establishment of a silent reservoir by Fc-FcR-mediated mechanisms. In established infection, bNAbs or bNAbs plus single inducers are ineffective in preventing viral rebound. However, bNAbs plus a combination of inducers that act by independent mechanisms synergize to decrease the reservoir as measured by viral rebound. Thus, combinations of inducers and bNAbs constitute a therapeutic strategy that impacts the establishment and maintenance of the HIV-1 reservoir in humanized mice.


Subject(s)
Antibodies, Neutralizing/administration & dosage , HIV Infections/immunology , HIV-1/drug effects , Transcription, Genetic/drug effects , Virus Latency/drug effects , Animals , Anti-HIV Agents/therapeutic use , Antibodies, Neutralizing/immunology , CD4-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/administration & dosage , HIV Infections/virology , HIV-1/genetics , HIV-1/physiology , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Humans , Hydroxamic Acids/administration & dosage , Immunoglobulin Fc Fragments/immunology , Mice , Receptors, Fc/immunology , Vorinostat
12.
Cancer Genomics Proteomics ; 10(4): 155-68, 2013.
Article in English | MEDLINE | ID: mdl-23893924

ABSTRACT

In order to overcome limitations of monoclonal antibodies, new protein-based scaffolds have been designed and evaluated pre-clinically, and some of them are in clinical studies for the treatment of cancer. These entities can be placed into two categories: scaffolds which bind ligands via amino acids in exposed loops and those in which ligand binding is mediated by amino acids in secondary structures, such as ß-sheet modules. Accordingly, we discuss adnectins, lipocalins, Kunitz domain-based binders, avimers, knottins, fynomers, atrimers and cytotoxic T-lymphocyte associated protein-4 (CTLA4)-based binders which fall into the first category, while darpins, affibodies, affilins and armadillo repeat protein-based scaffolds are members of the second category. In addition, we also discuss the new molecular entities as imaging tools and outline their unique characteristics in the context of multimeric and multivalent binding.


Subject(s)
Amino Acids/chemistry , Antibodies, Monoclonal/therapeutic use , Neoplasms/drug therapy , Proteins/administration & dosage , Amino Acids/therapeutic use , Antibodies, Monoclonal/chemistry , CTLA-4 Antigen/administration & dosage , CTLA-4 Antigen/chemistry , Humans , Ligands , Neoplasms/genetics , Neoplasms/immunology , Protein Binding , Protein Structure, Secondary , Proteins/chemistry
13.
J Immunol Methods ; 391(1-2): 103-11, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23470981

ABSTRACT

Targeted cell therapies are possible through the generation of recombinant fusion proteins that combine a toxin, such as diphtheria toxin (DT), with an antibody or other molecule that confers specificity. Upon binding of the fusion protein to the cell of interest, the diphtheria toxin is internalized which results in protein synthesis inhibition and subsequent cell death. We have recently expressed and purified the recombinant soluble porcine CTLA-4 both with and without N-glycosylation in yeast Pichia pastoris for in vivo use in our preclinical swine model. The glycosylated and non-N-glycosylated versions of this recombinant protein each bind to a porcine CD80 expressing B-cell lymphoma line (LCL13271) with equal affinity (K(D)=13 nM). In this study we have linked each of the glycosylated and non-N-glycosylated soluble porcine CTLA-4 proteins to the truncated diphtheria toxin DT390 through genetic engineering yielding three versions of the porcine CTLA-4 fusion toxins: 1) monovalent glycosylated soluble porcine CTLA-4 fusion toxin; 2) monovalent non-N-glycosylated soluble porcine CTLA-4 fusion toxin and 3) bivalent non-N-glycosylated soluble porcine CTLA-4 fusion toxin. Protein synthesis inhibition analysis demonstrated that while all three fusion toxins are capable of inhibiting protein synthesis in vitro, the non-N-glycosylated porcine CTLA-4 isoforms function most efficiently. Binding analysis using flow cytometry of the porcine CTLA-4 fusion toxins to LCL13271 cells also demonstrated that the non-N-glycosylated porcine CTLA-4 isoforms bind to these cells with higher affinity compared to the glycosylated fusion toxin. The monovalent non-N-glycosylated porcine CTLA-4 fusion toxin was tested in vivo. NSG (NOD/SCID IL-2 receptor γ(-)/(-)) mice were injected with porcine CD80(+) LCL13271 tumor cells. All animals succumbed to tumors and those treated with the monovalent non-N-glycosylated porcine CTLA-4 fusion toxin survived longer based on a symptomatic scoring system compared to the untreated controls. This recombinant protein may therefore provide a novel approach for in vivo depletion of porcine antigen presenting cells (APCs) for studies investigating the induction of transplantation tolerance, autoimmune disease and cancer treatment.


Subject(s)
CTLA-4 Antigen/administration & dosage , Diphtheria Toxin/administration & dosage , Immunotherapy/methods , Immunotoxins/administration & dosage , Lymphoma, B-Cell/therapy , Animals , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , CTLA-4 Antigen/biosynthesis , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Cell Line , Diphtheria Toxin/biosynthesis , Diphtheria Toxin/genetics , Diphtheria Toxin/metabolism , Flow Cytometry , Glycosylation , Immunotoxins/genetics , Immunotoxins/metabolism , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Pichia/genetics , Pichia/metabolism , Protein Binding , Protein Biosynthesis , Protein Processing, Post-Translational , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Swine
SELECTION OF CITATIONS
SEARCH DETAIL