Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Mol Cancer Ther ; 21(9): 1462-1472, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35793468

ABSTRACT

Extra domain B splice variant of fibronectin (EDB+FN) is an extracellular matrix protein (ECM) deposited by tumor-associated fibroblasts, and is associated with tumor growth, angiogenesis, and invasion. We hypothesized that EDB+FN is a safe and abundant target for therapeutic intervention with an antibody-drug conjugate (ADC). We describe the generation, pharmacology, mechanism of action, and safety profile of an ADC specific for EDB+FN (EDB-ADC). EDB+FN is broadly expressed in the stroma of pancreatic, non-small cell lung (NSCLC), breast, ovarian, head and neck cancers, whereas restricted in normal tissues. In patient-derived xenograft (PDX), cell-line xenograft (CLX), and mouse syngeneic tumor models, EDB-ADC, conjugated to auristatin Aur0101 through site-specific technology, demonstrated potent antitumor growth inhibition. Increased phospho-histone H3, a pharmacodynamic biomarker of response, was observed in tumor cells distal to the target site of tumor ECM after EDB-ADC treatment. EDB-ADC potentiated infiltration of immune cells, including CD3+ T lymphocytes into the tumor, providing rationale for the combination of EDB-ADC with immune checkpoint therapy. EDB-ADC and anti-PD-L1 combination in a syngeneic breast tumor model led to enhanced antitumor activity with sustained tumor regressions. In nonclinical safety studies in nonhuman primates, EDB-ADC had a well-tolerated safety profile without signs of either on-target toxicity or the off-target effects typically observed with ADCs that are conjugated through conventional conjugation methods. These data highlight the potential for EDB-ADC to specifically target the tumor microenvironment, provide robust therapeutic benefits against multiple tumor types, and enhance activity antitumor in combination with checkpoint blockade.


Subject(s)
Breast Neoplasms , Immunoconjugates , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Fibronectins/metabolism , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Mice , Neovascularization, Pathologic/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
2.
Clin Cancer Res ; 21(18): 4165-73, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26015513

ABSTRACT

PURPOSE: Triple-negative breast cancer (TNBC) and ovarian cancer each comprise heterogeneous tumors, for which current therapies have little clinical benefit. Novel therapies that target and eradicate tumor-initiating cells (TIC) are needed to significantly improve survival. EXPERIMENTAL DESIGN: A panel of well-annotated patient-derived xenografts (PDX) was established, and surface markers that enriched for TIC in specific tumor subtypes were empirically determined. The TICs were queried for overexpressed antigens, one of which was selected to be the target of an antibody-drug conjugate (ADC). The efficacy of the ADC was evaluated in 15 PDX models to generate hypotheses for patient stratification. RESULTS: We herein identified E-cadherin (CD324) as a surface antigen able to reproducibly enrich for TIC in well-annotated, low-passage TNBC and ovarian cancer PDXs. Gene expression analysis of TIC led to the identification of Ephrin-A4 (EFNA4) as a prospective therapeutic target. An ADC comprising a humanized anti-EFNA4 monoclonal antibody conjugated to the DNA-damaging agent calicheamicin achieved sustained tumor regressions in both TNBC and ovarian cancer PDX in vivo. Non-claudin low TNBC tumors exhibited higher expression and more robust responses than other breast cancer subtypes, suggesting a specific translational application for tumor subclassification. CONCLUSIONS: These findings demonstrate the potential of PF-06647263 (anti-EFNA4-ADC) as a first-in-class compound designed to eradicate TIC. The use of well-annotated PDX for drug discovery enabled the identification of a novel TIC target, pharmacologic evaluation of the compound, and translational studies to inform clinical development.


Subject(s)
Aminoglycosides/chemistry , Antibodies, Monoclonal, Murine-Derived/chemistry , Enediynes/chemistry , Ephrin-A4/chemistry , Ovarian Neoplasms/drug therapy , Triple Negative Breast Neoplasms/drug therapy , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antigens, Neoplasm/chemistry , Cell Line, Tumor , DNA/chemistry , Drug Design , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Prospective Studies , Random Allocation , Treatment Outcome , Xenograft Model Antitumor Assays
3.
Mol Cancer Ther ; 14(4): 952-63, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25646013

ABSTRACT

Antibody-drug conjugates (ADC) are emerging as clinically effective therapy. We hypothesized that cancers treated with ADCs would acquire resistance mechanisms unique to immunoconjugate therapy and that changing ADC components may overcome resistance. Breast cancer cell lines were exposed to multiple cycles of anti-Her2 trastuzumab-maytansinoid ADC (TM-ADC) at IC80 concentrations followed by recovery. The resistant cells, 361-TM and JIMT1-TM, were characterized by cytotoxicity, proteomic, transcriptional, and other profiling. Approximately 250-fold resistance to TM-ADC developed in 361-TM cells, and cross-resistance was observed to other non-cleavable-linked ADCs. Strikingly, these 361-TM cells retained sensitivity to ADCs containing cleavable mcValCitPABC-linked auristatins. In JIMT1-TM cells, 16-fold resistance to TM-ADC developed, with cross-resistance to other trastuzumab-ADCs. Both 361-TM and JIMT1-TM cells showed minimal resistance to unconjugated mertansine (DM1) and other chemotherapeutics. Proteomics and immunoblots detected increased ABCC1 (MRP1) drug efflux protein in 361-TM cells, and decreased Her2 (ErbB2) in JIMT1-TM cells. Proteomics also showed alterations in various pathways upon chronic exposure to the drug in both cell models. Tumors derived from 361-TM cells grew in mice and were refractory to TM-ADC compared with parental cells. Hence, acquired resistance to trastuzumab-maytansinoid ADC was generated in cultured cancer cells by chronic drug treatment, and either increased ABCC1 protein or reduced Her2 antigen were primary mediators of resistance. These ADC-resistant cell models retain sensitivity to other ADCs or standard-of-care chemotherapeutics, suggesting that alternate therapies may overcome acquired ADC resistance. Mol Cancer Ther; 14(4); 952-63. ©2015 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Immunoconjugates/pharmacology , Trastuzumab/pharmacology , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Immunoconjugates/administration & dosage , Inhibitory Concentration 50 , Mice , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Protein Transport , Proteome , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Signal Transduction , Transcriptome , Trastuzumab/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Mol Cancer Ther ; 12(1): 38-47, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23223830

ABSTRACT

Antibody-drug conjugates (ADC) represent a promising therapeutic modality for the clinical management of cancer. We sought to develop a novel ADC that targets 5T4, an oncofetal antigen expressed on tumor-initiating cells (TIC), which comprise the most aggressive cell population in the tumor. We optimized an anti-5T4 ADC (A1mcMMAF) by sulfydryl-based conjugation of the humanized A1 antibody to the tubulin inhibitor monomethylauristatin F (MMAF) via a maleimidocaproyl linker. A1mcMMAF exhibited potent in vivo antitumor activity in a variety of tumor models and induced long-term regressions for up to 100 days after the last dose. Strikingly, animals showed pathologic complete response in each model with doses as low as 3 mg antibody/kg dosed every 4 days. In a non-small cell lung cancer patient-derived xenograft model, in which 5T4 is preferentially expressed on the less differentiated tumor cells, A1mcMMAF treatment resulted in sustained tumor regressions and reduced TIC frequency. These results highlight the potential of ADCs that target the most aggressive cell populations within tumors, such as TICs. In exploratory safety studies, A1mcMMAF exhibited no overt toxicities when administered to cynomolgus monkeys at doses up to 10 mg antibody/kg/cycle × 2 and displayed a half-life of 5 days. The preclinical efficacy and safety data established a promising therapeutic index that supports clinical testing of A1mcMMAF.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Membrane Glycoproteins/metabolism , Neoplastic Stem Cells/metabolism , Animals , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal, Humanized/toxicity , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Humans , Inhibitory Concentration 50 , Macaca fascicularis , Male , Maximum Tolerated Dose , Membrane Glycoproteins/immunology , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Remission Induction , Tissue Distribution , Tubulin Modulators/chemical synthesis , Tubulin Modulators/pharmacology , Xenograft Model Antitumor Assays
5.
Cancer Res ; 71(12): 4236-46, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21540235

ABSTRACT

Poorly differentiated tumors in non-small cell lung cancer (NSCLC) have been associated with shorter patient survival and shorter time to recurrence following treatment. Here, we integrate multiple experimental models with clinicopathologic analysis of patient tumors to delineate a cellular hierarchy in NSCLC. We show that the oncofetal protein 5T4 is expressed on tumor-initiating cells and associated with worse clinical outcome in NSCLC. Coexpression of 5T4 and factors involved in the epithelial-to-mesenchymal transition were observed in undifferentiated but not in differentiated tumor cells. Despite heterogeneous expression of 5T4 in NSCLC patient-derived xenografts, treatment with an anti-5T4 antibody-drug conjugate resulted in complete and sustained tumor regression. Thus, the aggressive growth of heterogeneous solid tumors can be blocked by therapeutic agents that target a subpopulation of cells near the top of the cellular hierarchy.


Subject(s)
Antigens, Neoplasm/analysis , Carcinoma, Non-Small-Cell Lung/drug therapy , Immunotoxins/therapeutic use , Lung Neoplasms/drug therapy , Membrane Glycoproteins/analysis , Neoplastic Stem Cells/immunology , Animals , CD24 Antigen/analysis , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Humans , Hyaluronan Receptors/analysis , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Membrane Glycoproteins/physiology , Mice
6.
Cancer Chemother Pharmacol ; 61(6): 1027-35, 2008 May.
Article in English | MEDLINE | ID: mdl-17668210

ABSTRACT

PURPOSE: The present study aims to establish a method that provides fast, precise and reproducible pharmacokinetic (PK) parameters of antibody-calicheamicin conjugates. The method should discriminate between PK of the antibody moiety and PK of the conjugated calicheamicin (CM). METHODS: The conjugates gemtuzumab ozogamicin (CMA-676, Mylotarg) or inotuzumab ozogamicin (CMC-544) were injected in the tail vein of nude mice. At regular time intervals, 5 mul whole blood samples were taken from the tail artery. Concentrations of conjugated CMA-676 or CMC-544 as well as concentrations of their respective antibody moiety were determined by sandwich plasmon resonance. This detection system measures changes in the plasma resonance angle caused by the interaction of macromolecules on biosensor chips. We determined as a first measure the binding of CMA-676 or CMC-544 to their respective antigens, CD33 or CD22. As a second measure we determined the amount of CM on the antigen-bound conjugates. This was done by determination of changes in plasma resonance angle after binding of an anti-CM antibody. RESULTS: Sandwich plasmon resonance allowed detection of both conjugates in blood of mice in a range of 100-1,000 ng/ml protein. Due to the precision of the sampling and detection methods, PK values of each conjugate were determined in individual mice. Calicheamicin bound to antibody was eliminated faster than the antibody alone. The presence of a CD22-expressing tumour in mice reduced the plasma levels of the CD22-targeting conjugate but not of the CD33-targeting one. CONCLUSIONS: Using small blood samples from a mouse, the sandwich plasmon resonance method provided PK-values of CM-conjugates and information about the stability of the linkage in vivo. Comparison between the PK-values of CM-conjugates in tumour-bearing and tumour-free mice suggested that retention of the conjugate in tumour tissue due to antigen targeting could be deduced from the plasma levels.


Subject(s)
Aminoglycosides/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Aminoglycosides/administration & dosage , Aminoglycosides/blood , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/blood , Antibodies, Monoclonal, Humanized , Area Under Curve , Cell Line, Tumor , Gemtuzumab , Half-Life , Humans , Injections, Intraperitoneal , Injections, Intravenous , Inotuzumab Ozogamicin , Mice , Mice, Inbred BALB C , Mice, Nude , Rabbits , Surface Plasmon Resonance
7.
Int J Oncol ; 28(3): 675-84, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16465373

ABSTRACT

Calicheamicin is a potent chemotherapeutic with a low therapeutic index that requires targeting to tumor cells for its use in the clinic. To treat acute myeloid leukemia, calicheamicin has been conjugated to an antibody that recognizes CD33 (gemtuzumab ozogamicin). The application range of this 'active' targeting strategy is limited since it depends on specific antigen expression by tumor cells. This limitation could be reduced by using an antigen-independent 'passive targeting' strategy for calicheamicin. 'Passive targeting' relies on the dysfunctional vasculature of a neoplastic tumor that allows enhanced retention of macromolecules. We studied the efficacy of calicheamicin conjugated to various carrier molecules: i.e. immunoglobulin, albumin or PEGylated Fc fragments. In nude mice, a conjugate of anti-CD33 and calicheamicin accumulates in human tumor xenografts in the absence of detectable amounts of targeting antigen. Passive targeting provided sufficient accumulation of this conjugate to inhibit tumor growth of 10 different CD33-negative xenograft models. This efficacy depended on the use of an acid-labile linker between antibody and calicheamicin. Substitution of immunoglobulin as a carrier with either albumin or PEGylated Fc reduced or eliminated the efficacy of the conjugate. The results showed that using 'non-specific' immunoglobulin for passive targeting of calicheamicin might be an effective mode of cancer therapy.


Subject(s)
Aminoglycosides/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Immunoconjugates/therapeutic use , Xenograft Model Antitumor Assays/methods , Aminoglycosides/pharmacokinetics , Aminoglycosides/pharmacology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Female , Gemtuzumab , HT29 Cells , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/therapeutic use , Inhibitory Concentration 50 , Male , Mice , Mice, Nude , Polyethylene Glycols/chemistry , Rituximab , Serum Albumin/therapeutic use
8.
Cancer Immunol Immunother ; 54(1): 11-24, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15693135

ABSTRACT

Antibody-targeted chemotherapy with immunoconjugates of calicheamicin is a clinically validated strategy in cancer therapy. This study describes the selection of a murine anti-CD22 mAb, m5/44, as a targeting agent, its conjugation to a derivative of calicheamicin (CalichDM) via either acid-labile or acid-stable linkers, the antitumor activity of CalichDM conjugated to m5/44, and its subsequent humanization by CDR grafting. Murine IgG1 mAb m5/44 was selected based on its subnanomolar affinity for CD22 and ability to be internalized into B cells. CalichDM conjugated to m5/44 caused potent growth inhibition of CD22+ human B-cell lymphomas (BCLs) in vitro. The conjugate of m5/44 with an acid-labile linker was more potent than an acid-stable conjugate, a nonbinding conjugate with a similar acid-labile linker, or unconjugated CalichDMH in inhibiting BCL growth. CalichDM conjugated to m5/44 caused regression of established BCL xenografts in nude mice. In contrast, both unconjugated m5/44 and a nonbinding conjugate were ineffective against these xenografts. Based on the potent antitumor activity of m5/44-CalichDM conjugates, m5/44 was humanized by CDR grafting to create g5/44, an IgG4 anti-CD22 antibody. Both m5/44 and g5/44 bound CD22 with subnanomolar affinity. Competitive blocking with previously characterized murine anti-CD22 mAbs suggested that g5/44 recognizes epitope A located within the first N-terminal Ig-like domain of human CD22. Antitumor efficacy of CalichDM conjugated to g5/44 against BCL xenografts was more potent than its murine counterpart. Based on these results, a calicheamicin conjugate of g5/44, CMC-544, was selected for further development as a targeted chemotherapeutic agent for the treatment of B-cell malignancies.


Subject(s)
Aminoglycosides/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Antineoplastic Agents/therapeutic use , Cell Adhesion Molecules/immunology , Immunoconjugates/therapeutic use , Lectins/immunology , Lymphoma, B-Cell/therapy , Amino Acid Sequence , Aminoglycosides/chemistry , Aminoglycosides/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antineoplastic Agents/immunology , Binding, Competitive , Cell Line, Tumor , Epitopes/immunology , Female , Humans , Immunoconjugates/immunology , Lymphoma, B-Cell/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Sialic Acid Binding Ig-like Lectin 2 , Xenograft Model Antitumor Assays/methods
9.
Clin Cancer Res ; 10(13): 4538-49, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15240546

ABSTRACT

PURPOSE: Linking a cytotoxic anticancer drug to an antibody that recognizes a tumor-associated antigen can improve the therapeutic index of the drug. We asked whether a conjugate of the cytotoxic antibiotic N-acetyl gamma calicheamicin dimethyl hydrazide (CalichDMH) and an antibody recognizing Lewis(y) (Le(y)) antigen could eliminate carcinomas that express Le(y). Because Le(y) is highly expressed on carcinomas of colon, breast, lung, ovary, and prostate, a CalichDMH conjugate targeting Le(y) could provide a treatment option for various cancers. EXPERIMENTAL DESIGN: The humanized anti-Le(y) antibody hu3S193 was conjugated to CalichDMH via the bifunctional AcBut linker. Selectivity and avidity of the conjugate (hu3S193-CalichDMH) for Le(y)-BSA or Le(y+) cells was tested by BIAcore or flow cytometry. Cytotoxicity of hu3S193-CalichDMH was compared with toxicity of a control conjugate on monolayers of Le(y+) and Le(y-) carcinoma cells. Inhibition of tumor growth by hu3S193-CalichDMH was assessed on three types of s.c. xenografts. RESULTS: Hu3S193-CalichDMH had similar selectivity as hu3S193. The conjugate had lower affinity for Le(y)-BSA but not for Le(y+) cells. When tested on monolayers of human Le(y+) carcinoma cells, hu3S193-CalichDMH was more cytotoxic than a control conjugate. This difference in efficacy was not noted on Le(y-) cells. Efficacy of hu3S193-CalichDMH depended on the expression of Le(y) and on the sensitivity of the cells to CalichDMH. In vivo, hu3S193-CalichDMH inhibited growth of xenografted human gastric (N87), colon (LOVO), and prostate carcinomas (LNCaP). When used against N87 xenografts, hu3S193-CalichDMH arrested tumor growth for at least 100 days. CONCLUSION: Hu3S193-CalichDMH can specifically eliminate Le(y+) tumors. These results support development of this conjugate for treatment of carcinomas.


Subject(s)
Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Hydrazines/pharmacology , Immunotherapy/methods , Lewis Blood Group Antigens/chemistry , Animals , Antigens/chemistry , Carcinoma/metabolism , Cell Line, Tumor , Cell Separation , Collagen/chemistry , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Drug Combinations , Enediynes , Female , Flow Cytometry , Humans , Hydrolysis , Kinetics , Laminin/chemistry , Male , Mice , Mice, Nude , Models, Chemical , Neoplasm Transplantation , Protein Binding , Proteoglycans/chemistry , Sensitivity and Specificity , Surface Plasmon Resonance , Tissue Distribution
10.
Blood ; 103(5): 1807-14, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14615373

ABSTRACT

Antibody-targeted chemotherapy with gemtuzumab ozogamicin (CMA-676, a CD33-targeted immunoconjugate of N-acetyl-gamma-calicheamicin dimethyl hydrazide [CalichDMH], a potent DNA-binding cytotoxic antitumor antibiotic) is a clinically validated therapeutic option for patients with acute myeloid leukemia (AML). Here, we describe the preclinical profile of another immunoconjugate of CalichDMH, CMC-544, targeted to CD22 expressed by B-lymphoid malignancies. CMC-544 comprises a humanized IgG4 anti-CD22 monoclonal antibody (mAb), G5/44, covalently linked to CalichDMH via an acid-labile 4-(4'-acetylphenoxy) butanoic acid (AcBut) linker. Both CMC-544 and unconjugated G5/44 bound human CD22 with subnanomolar affinity. CMC-544, but not unconjugated G5/44, exerted potent cytotoxicity against CD22+ B-cell lymphoma (BCL) cell lines (inhibitory concentration of 50%: 6-600 pM CalichDMH). CMC-544 caused a potent inhibition of growth of small but established BCL xenografts leading to cures (therapeutic index > 10). CMC-544 prevented the establishment of BCL xenografts and also caused regression of large BCLs (> 1.5 g tumor mass). In contrast, unconjugated CalichDMH, unconjugated G5/44, and an isotype-matched control conjugate, CMA-676, were ineffective against these BCL xenografts. Thus, CD22-targeted delivery of CalichDMH is a potent and effective preclinical therapeutic strategy for BCLs. The strong antitumor profile of CMC-544 supports its clinical evaluation as a treatment option for B-lymphoid malignancies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/biosynthesis , Antigens, Differentiation, B-Lymphocyte/biosynthesis , Cell Adhesion Molecules , Immunoconjugates/therapeutic use , Lectins/biosynthesis , Lymphoma, B-Cell/therapy , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , Immunoglobulin G/metabolism , Immunotherapy/methods , Inhibitory Concentration 50 , Inotuzumab Ozogamicin , Lectins/metabolism , Lymphoma, B-Cell/metabolism , Lymphoma, Non-Hodgkin/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Chemical , Neoplasm Transplantation , Protein Binding , Sialic Acid Binding Ig-like Lectin 2 , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...