Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Bioorg Med Chem ; 28(23): 115785, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33099182

ABSTRACT

ADCs based on the natural product maytansine have been successfully employed clinically. In a previous report, ADCs based on hydrophilic non-cell permeable maytansinoids was presented. The authors in this report further explore the maytansine scaffold to develop tubulin inhibitors capable of cell permeation. The research resulted in amino-benzoyl-maytansinoid payloads that were further elaborated with linkers for conjugating to antibodies. This approach was applied to MUC16 tumor targeting antibodies for ovarian cancers. A positive control ADC was evaluated alongside the amino-benzoyl-maytansinoid ADC and the efficacy observed was equivalent while the isotype control ADCs had no effect.


Subject(s)
Immunoconjugates/metabolism , Maytansine/chemistry , Tubulin Modulators/chemistry , Animals , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Female , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Maytansine/metabolism , Mice, SCID , Neoplasms/drug therapy , Neoplasms/pathology , Structure-Activity Relationship , Transplantation, Heterologous , Tubulin Modulators/metabolism
2.
Bioorg Med Chem ; 26(9): 2271-2279, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29605304

ABSTRACT

Natural products have been used for many medicinal purposes for centuries. Antibody drug conjugates (ADCs) have utilized this rich source of small molecule therapeutics to produce several clinically useful treatments. ADCs based on the natural product maytansine have been successful clinically. The authors further the utility of the anti-cancer natural product maytansine by developing efficacious payloads and linker-payloads for conjugating to antibodies. The success of our approach was realized in the EGFRvIII targeting ADC EGFRvIII-16. The ADC was able to regress tumors in 2 tumor models (U251/EGFRvIII and MMT/EGFRvIII). When compared to a positive control ADC, the efficacy observed was similar or improved while the isotype control ADCs had no effect.


Subject(s)
Antineoplastic Agents/pharmacology , Immunotoxins/pharmacology , Maytansine/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , CHO Cells , Cell Line, Tumor , Cricetulus , ErbB Receptors/immunology , Female , Humans , Hydrophobic and Hydrophilic Interactions , Immunotoxins/chemistry , Immunotoxins/immunology , Kinetics , Male , Maytansine/chemical synthesis , Maytansine/chemistry , Mice , Xenograft Model Antitumor Assays
3.
Mol Cancer Ther ; 20(10): 1966-1976, 2021 10.
Article in English | MEDLINE | ID: mdl-34315762

ABSTRACT

Lung cancers harboring mesenchymal-to-epithelial transition factor (MET) genetic alterations, such as exon 14 skipping mutations or high-level gene amplification, respond well to MET-selective tyrosine kinase inhibitors (TKI). However, these agents benefit a relatively small group of patients (4%-5% of lung cancers), and acquired resistance limits response durability. An antibody-drug conjugate (ADC) targeting MET might enable effective treatment of MET-overexpressing tumors (approximately 25% of lung cancers) that do not respond to MET targeted therapies. Using a protease-cleavable linker, we conjugated a biparatopic METxMET antibody to a maytansinoid payload to generate a MET ADC (METxMET-M114). METxMET-M114 promotes substantial and durable tumor regression in xenografts with moderate to high MET expression, including models that exhibit innate or acquired resistance to MET blockers. Positron emission tomography (PET) studies show that tumor uptake of radiolabeled METxMET antibody correlates with MET expression levels and METxMET-M114 efficacy. In a cynomolgus monkey toxicology study, METxMET-M114 was well tolerated at a dose that provides circulating drug concentrations that are sufficient for maximal antitumor activity in mouse models. Our findings suggest that METxMET-M114, which takes advantage of the unique trafficking properties of our METxMET antibody, is a promising candidate for the treatment of MET-overexpressing tumors, with the potential to address some of the limitations faced by the MET function blockers currently in clinical use.


Subject(s)
Antibodies, Monoclonal/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , Immunoconjugates/pharmacology , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Female , Humans , Immunoconjugates/pharmacokinetics , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Macaca fascicularis , Male , Mice , Mice, SCID , Mutation , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
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
5.
Mol Cancer Ther ; 16(4): 681-693, 2017 04.
Article in English | MEDLINE | ID: mdl-28108597

ABSTRACT

The properties of cell surface proteins targeted by antibody-drug conjugates (ADCs) have not been fully exploited; of particular importance are the rate of internalization and the route of intracellular trafficking. In this study, we compared the trafficking of HER2, which is the target of the clinically approved ADC ado-trastuzumab emtansine (T-DM1), with that of prolactin receptor (PRLR), another potential target in breast cancer. In contrast to HER2, we found that PRLR is rapidly and constitutively internalized, and traffics efficiently to lysosomes, where it is degraded. The PRLR cytoplasmic domain is necessary to promote rapid internalization and degradation, and when transferred to HER2, enhances HER2 degradation. In accordance with these findings, low levels of cell surface PRLR (∼30,000 surface receptors per cell) are sufficient to mediate effective killing by PRLR ADC, whereas cell killing by HER2 ADC requires higher levels of cell surface HER2 (∼106 surface receptors per cell). Noncovalently cross-linking HER2 to PRLR at the cell surface, using a bispecific antibody that binds to both receptors, dramatically enhances the degradation of HER2 as well as the cell killing activity of a noncompeting HER2 ADC. Furthermore, in breast cancer cells that coexpress HER2 and PRLR, a HER2xPRLR bispecific ADC kills more effectively than HER2 ADC. These results emphasize that intracellular trafficking of ADC targets is a key property for their activity and, further, that coupling an ADC target to a rapidly internalizing protein may be a useful approach to enhance internalization and cell killing activity of ADCs. Mol Cancer Ther; 16(4); 681-93. ©2017 AACR.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/metabolism , Immunoconjugates/pharmacology , Maytansine/analogs & derivatives , Receptor, ErbB-2/antagonists & inhibitors , Receptors, Prolactin/antagonists & inhibitors , Ado-Trastuzumab Emtansine , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Female , Humans , Maytansine/pharmacology , Protein Transport/drug effects , Receptor, ErbB-2/metabolism , Receptors, Prolactin/metabolism , Trastuzumab
6.
Mol Cancer Ther ; 16(7): 1299-1311, 2017 07.
Article in English | MEDLINE | ID: mdl-28377489

ABSTRACT

The Prolactin Receptor (PRLR) is a type 1 cytokine receptor that is expressed in a subset of breast cancers and may contribute to its pathogenesis. It is relatively overexpressed in approximately 25% of human breast tumors while expressed at low levels in some normal human tissues including the mammary gland. We developed an anti-PRLR antibody-drug conjugate (ADC), to target PRLR-positive breast cancer. REGN2878-DM1 is comprised of a fully human high-affinity function-blocking anti-PRLR IgG1 antibody (REGN2878) conjugated via a noncleavable SMCC linker to the cytotoxic maytansine derivative DM1. Both unconjugated REGN2878 and conjugated REGN2878-DM1 block PRL-mediated activation in vitro and are rapidly internalized into lysosomes. REGN2878-DM1 induces potent cell-cycle arrest and cytotoxicity in PRLR-expressing tumor cell lines. In vivo, REGN2878-DM1 demonstrated significant antigen-specific antitumor activity against breast cancer xenograft models. In addition, REGN2878-DM1 showed additive activity when combined with the antiestrogen agent fulvestrant. These results illustrate promising antitumor activity against PRLR-positive breast cancer xenografts and support the evaluation of anti-PRLR ADCs as potential therapeutic agents in breast cancer. Mol Cancer Ther; 16(7); 1299-311. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Breast Neoplasms/drug therapy , Immunoconjugates/administration & dosage , Receptors, Prolactin/immunology , Animals , Antibodies, Anti-Idiotypic/administration & dosage , Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal, Humanized/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Female , Humans , Immunoconjugates/immunology , Mice , Receptors, Prolactin/genetics , Xenograft Model Antitumor Assays
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.
Clin Cancer Res ; 10(24): 8620-9, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15623646

ABSTRACT

PURPOSE: CMC-544 is a CD22-targeted immunoconjugate of calicheamicin and exerts a potent cytotoxic effect against CD22+ B-cell lymphoma. This study evaluated antitumor efficacy of CMC-544 against systemically disseminated B-cell lymphoma. EXPERIMENTAL DESIGN: Scid mice received i.v. injections of CD22+ Ramos B-cell lymphoma cells for their systemic dissemination. CMC-544, G5/44, CD33-targeted CMA-676 (control conjugate) or rituximab were given i.p. 3, 9, 15, or 21 days after B-cell lymphoma dissemination. Diseased mice were monitored daily for hind-limb paralysis and death. Histopathological examination of CMC-544-treated and vehicle-treated diseased mice was also performed. RESULTS: Mice with disseminated B-cell lymphoma developed hind-limb paralysis within 35 days. When given up to 15 days after B-cell lymphoma dissemination, CMC-544 extended survival of the diseased mice to >100 days, and these mice were considered cured. CMC-544 was efficacious when given during both the early initiation phase and the late established phase of the disease. A single dose of CMC-544 was effective in delaying the occurrence of hind-limb paralysis. In contrast, neither CMA-676 nor unconjugated G5/44 was effective. Rituximab was effective when given early in the disease process but not when the disease was established. Histopathological analysis revealed B-cell lymphoma infiltration in brain, spinal cord, bone marrow, and kidney in vehicle-treated but not in CMC-544-treated diseased mice. Consistent with its efficacy against the disseminated B-cell lymphoma, CMC-544 also caused regression of established Ramos B-cell lymphoma xenografts in scid mice. CONCLUSIONS: CMC-544 confers strong therapeutic activity against systemic disseminated B-cell lymphoma and protects mice from hind-limb paralysis and death. These results support clinical evaluation of CMC-544 in the treatment of CD22+ lymphoid malignancies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , Cell Adhesion Molecules/metabolism , Hindlimb , Immunoconjugates/therapeutic use , Lectins/metabolism , Lymphoma, B-Cell/therapy , Paralysis/etiology , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Humans , Immunoglobulin G/metabolism , Immunotherapy/methods , Inotuzumab Ozogamicin , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Male , Mice , Mice, SCID , Rituximab , Sialic Acid Binding Ig-like Lectin 2 , Survival Rate , Transplantation, Heterologous
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.
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
11.
Bioconjug Chem ; 19(1): 358-61, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17994681

ABSTRACT

The formation of protein aggregates can be a major problem during the preparation of antibody-drug conjugates. Herein is described the methods by which reaction additives were selected, which reduce the tendency of antibodies to aggregate during the attachment of the cytotoxic agent calicheamicin to form an immunoconjugate. Reaction conditions were delineated that produced optimized yields of monomeric conjugates. These conditions were used in the preclinical preparations of gemtuzumab ozogamicin (Mylotarg), the first commercially available chemotherapeutic immunoconjugate.


Subject(s)
Aminoglycosides/chemical synthesis , Aminoglycosides/immunology , Antibodies, Monoclonal/immunology , Immunotoxins/chemistry , Aminoglycosides/chemistry , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Gemtuzumab , Humans , Immunoglobulin G/immunology , Immunotoxins/immunology , Organic Chemicals/chemistry , Sialic Acid Binding Ig-like Lectin 3 , United States , United States Food and Drug Administration
12.
Cancer Immunol Immunother ; 56(7): 1107-17, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17160682

ABSTRACT

Tumor-targeted delivery of a potent cytotoxic agent, calicheamicin, using its immunoconjugates is a clinically validated therapeutic strategy. Rituximab is a human CD20-specific chimeric antibody extensively used in B-NHL therapy. We investigated whether conjugation to calicheamicin can improve the anti-tumor activity of rituximab against human B-cell lymphoma (BCL) xenografts in preclinical models. BCL cells were cultured with rituximab or its calicheamicin conjugates and their in vitro growth was monitored. BCL cells were injected s.c. to establish localized xenografts in nude mice or i.v. to establish disseminated BCL in severe combined immunodeficient (scid) mice. I.p. treatment with rituximab or its calicheamicin conjugates was initiated and its effect on s.c. BCL growth or survival of mice with disseminated BCL was monitored. Conjugation of calicheamicin to rituximab vastly enhanced its growth inhibitory activity against BCL in vitro. Conjugation to calicheamicin had no deleterious effect on the effector functional activity of rituximab. Calicheamicin conjugated to rituximab with an acid-labile linker exhibited greater anti-tumor activity against s.c. BCL xenografts and improved survival of mice with disseminated BCL over that of unconjugated rituximab. Anti-tumor activities of rituximab conjugated to calicheamicin via an acid-stable linker were similar to that of unconjugated rituximab. Superior anti-tumor efficacy exhibited by a calicheamicin immunoconjugate of rituximab with an acid-labile linker over that of rituximab demonstrates the therapeutic potential of CD20-specific antibody-targeted chemotherapy strategy in the treatment of B-NHL.


Subject(s)
Aminoglycosides/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antigens, CD20/immunology , Drug Delivery Systems/methods , Enediynes/administration & dosage , Immunoconjugates/administration & dosage , Lymphoma, B-Cell/drug therapy , Aminoglycosides/immunology , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Murine-Derived , Antibody Specificity , Cell Proliferation/drug effects , Cytotoxicity, Immunologic/drug effects , Enediynes/immunology , Flow Cytometry , Humans , Immunoconjugates/immunology , Lymphoma, B-Cell/immunology , Mice , Mice, Nude , Mice, SCID , Rituximab
13.
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
14.
Blood ; 106(7): 2235-43, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-15947095

ABSTRACT

We report the development of a mouse B cell-depleting immunoconjugate (anti-CD22 monoclonal antibody [mAb] conjugated to calicheamicin) and its in vivo use to characterize the kinetics of CD22+ B-cell depletion and reconstitution in murine primary and secondary lymphoid tissues. The effect of B-cell depletion was further studied in a murine collagen-induced arthritis (CIA) model and a respiratory syncytial virus (RSV) vaccination model. Our results show that (1) the immunoconjugate has B-cell-specific in vitro and in vivo cytotoxicity; (2) B-cell reconstitution starts in the bone marrow and spleen around day 30 after depletion and is completed in all tissues tested by day 50; (3) B-cell depletion inhibits the development of clinical and histologic arthritis in the CIA model; (4) depletion of type II collagen antibody levels is not necessary for clinical and histologic prevention of CIA; and (5) B-cell depletion does not adversely affect memory antibody responses after challenge nor clearance of infectious virus from lungs in the RSV vaccination model. These results demonstrate for the first time that only B-cell reduction but not type II collagen antibody levels correlate with the prevention of arthritis and represent key insights into the role of CD22-targeted B-cell depletion in mouse autoimmunity and vaccination models.


Subject(s)
Arthritis, Experimental/therapy , B-Lymphocytes/cytology , Animals , Arthritis, Experimental/metabolism , Autoimmunity , B-Lymphocytes/metabolism , Cell Proliferation , Collagen Type II/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoglobulin M/blood , Immunologic Memory , Inflammation , Kinetics , Male , Mice , Mice, Inbred C57BL , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Viruses/genetics , T-Lymphocytes/cytology , Time Factors
15.
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