Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Science ; 371(6526): 300-305, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33446559

ABSTRACT

Signaling through the CD19-CD81 co-receptor complex, in combination with the B cell receptor, is a critical determinant of B cell development and activation. It is unknown how CD81 engages CD19 to enable co-receptor function. Here, we report a 3.8-angstrom structure of the CD19-CD81 complex bound to a therapeutic antigen-binding fragment, determined by cryo-electron microscopy (cryo-EM). The structure includes both the extracellular domains and the transmembrane helices of the complex, revealing a contact interface between the ectodomains that drives complex formation. Upon binding to CD19, CD81 opens its ectodomain to expose a hydrophobic CD19-binding surface and reorganizes its transmembrane helices to occlude a cholesterol binding pocket present in the apoprotein. Our data reveal the structural basis for CD19-CD81 complex assembly, providing a foundation for rational design of therapies for B cell dysfunction.


Subject(s)
Antigens, CD19/chemistry , Receptors, Antigen, B-Cell/chemistry , Tetraspanin 28/chemistry , Amino Acid Sequence , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antigens, CD19/immunology , B-Lymphocytes/immunology , Cryoelectron Microscopy , Humans , Maytansine/analogs & derivatives , Maytansine/chemistry , Maytansine/immunology , Models, Molecular , Mutation , Protein Binding , Protein Domains , Receptors, Antigen, B-Cell/immunology , Tetraspanin 28/genetics , Tetraspanin 28/immunology
2.
Mol Pharm ; 16(9): 3926-3937, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31287952

ABSTRACT

Antibody-drug conjugates are an emerging class of cancer therapeutics constructed from monoclonal antibodies conjugated with small molecule effectors. First-generation molecules of this class often employed heterogeneous conjugation chemistry, but many site-specifically conjugated ADCs have been described recently. Here, we undertake a systematic comparison of ADCs made with the same antibody and the same macrocyclic maytansinoid effector but conjugated either heterogeneously at lysine residues or site-specifically at cysteine residues. Characterization of these ADCs in vitro reveals generally similar properties, including a similar catabolite profile, a key element in making a meaningful comparison of conjugation chemistries. In a mouse model of cervical cancer, the lysine-conjugated ADC affords greater efficacy on a molar payload basis. Rather than making general conclusions about ADCs conjugated by a particular chemistry, we interpret these results as highlighting the complexity of ADCs and the interplay between payload class, linker chemistry, target antigen, and other variables that determine efficacy in a given setting.


Subject(s)
Antibodies, Monoclonal/chemistry , Cysteine/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Lysine/chemistry , Maytansine/immunology , Uterine Cervical Neoplasms/drug therapy , Animals , Cell Survival/drug effects , Female , HeLa Cells , Humans , Immunoconjugates/administration & dosage , Injections, Intravenous , Mice , Mice, SCID , Treatment Outcome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
Clin Cancer Res ; 23(19): 5836-5845, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28679774

ABSTRACT

Purpose: Carcinosarcomas (CS) are highly aggressive gynecologic malignancies containing both carcinomatous and sarcomatous elements with heterogeneous HER2/neu expression. We compared the efficacy of SYD985 (Synthon Biopharmaceuticals BV), a novel HER2-targeting antibody-drug conjugate (ADC), to trastuzumab emtansine (T-DM1, Genentech-Roche) against primary uterine and ovarian CS.Experimental Design: Eight primary CS cell lines were evaluated for HER2/neu surface expression by IHC and gene amplification by FISH assays. The in vitro experiments included cytotoxicity, antibody-dependent cellular cytotoxicity (ADCC), proliferation, viability, and bystander killing. In vivo activity was studied in mouse xenograft and patient-derived xenograft (PDX) models.Results: SYD985 and T-DM1 induced similar levels of ADCC against CS cell lines with low and high HER2/neu expression when challanged in the presence of effector cells. In contrast, SYD985 was 7- to 54-fold more potent than T-DM1 in the absence of effector cells. SYD985, unlike T-DM1, was active against CS demonstrating low or heterogeneous HER2/neu expression. Specifically, the mean IC50 values were 0.060 µg/mL and 3.221 µg/mL (P < 0.0001) against HER2/neu 0/1+ cell lines and 0.013 µg/mL and 0.096 µg/mL (P < 0.0001) against HER2/neu 3+ cell lines for SYD985 versus T-DM1, respectively. Importantly, unlike T-DM1, SYD985 induced efficient bystander killing of HER2/neu 0/1+ tumor cells admixed with HER2/neu 3+ cells. In vivo studies confirmed that SYD985 is more active than T-DM1 in CS and highly effective against HER2/neu expressing xenografts and PDX.Conclusions: SYD985 may represent a novel and highly effective ADC against HER2-expressing CS. Clinical studies with SYD985 in patients harboring chemotherapy-resistant CS with low/moderate and high HER2 expression are warranted. Clin Cancer Res; 23(19); 5836-45. ©2017 AACR.


Subject(s)
Carcinosarcoma/drug therapy , Immunoconjugates/administration & dosage , Ovarian Neoplasms/drug therapy , Receptor, ErbB-2/genetics , Uterine Neoplasms/drug therapy , Ado-Trastuzumab Emtansine , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Carcinosarcoma/genetics , Carcinosarcoma/immunology , Carcinosarcoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Duocarmycins , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoconjugates/immunology , Indoles/administration & dosage , Indoles/immunology , Maytansine/administration & dosage , Maytansine/analogs & derivatives , Maytansine/immunology , Mice , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Pyrrolidinones/administration & dosage , Pyrrolidinones/immunology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/immunology , Trastuzumab/administration & dosage , Trastuzumab/immunology , Uterine Neoplasms/genetics , Uterine Neoplasms/immunology , Uterine Neoplasms/pathology , Xenograft Model Antitumor Assays
4.
AAPS J ; 19(4): 1223-1234, 2017 07.
Article in English | MEDLINE | ID: mdl-28534292

ABSTRACT

Antibody-drug-conjugates (ADCs) are an emerging class of biological therapeutics. Mirvetuximab soravtansine is a novel folate receptor alpha (FRα)-targeting ADC which represents a potential new treatment for patients with ovarian and other FRα-positive cancers. Since patient immune responses to biological therapeutics may negatively affect drug efficacy and patient safety, regulatory authorities require rigorous monitoring of patient samples. Taking advantage of the immune system's ability to generate highly specific antibodies, the field has turned to anti-idiotype antibodies as powerful tools for the development of sensitive and specific bioassays. Here, we report the generation and characterization of a highly specific neutralizing anti-idiotype antibody directed against M9346A, the antibody moiety of mirvetuximab soravtansine. The anti-idiotype antibody recognizes M9346A with double-digit picomolar affinity, competes with folate receptor antigen for binding to M9346A, and can be used to develop both anti-drug-antibody and neutralizing antibody assays.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Neutralizing/immunology , Immunoconjugates/immunology , Maytansine/analogs & derivatives , Animals , Humans , Maytansine/immunology , Mice
5.
Mol Cancer Ther ; 16(5): 879-892, 2017 05.
Article in English | MEDLINE | ID: mdl-28258164

ABSTRACT

Antibody-drug conjugates (ADC) are highly potent and specific antitumor drugs, combining the specific targeting of mAbs with the potency of small-molecule toxic payloads. ADCs generated by conventional chemical conjugation yield heterogeneous mixtures with variable pharmacokinetics, stability, safety, and efficacy profiles. To address these issues, numerous site-specific conjugation technologies are currently being developed allowing the manufacturing of homogeneous ADCs with predetermined drug-to-antibody ratios. Here, we used sortase-mediated antibody conjugation (SMAC) technology to generate homogeneous ADCs based on a derivative of the highly potent anthracycline toxin PNU-159682 and a noncleavable peptide linker, using the anti-HER2 antibody trastuzumab (part of Kadcyla) and the anti-CD30 antibody cAC10 (part of Adcetris). Characterization of the resulting ADCs in vitro and in vivo showed that they were highly stable and exhibited potencies exceeding those of ADCs based on conventional tubulin-targeting payloads, such as Kadcyla and Adcetris. The data presented here suggest that such novel and highly potent ADC formats may help to increase the number of targets available to ADC approaches, by reducing the threshold levels of target expression required. Mol Cancer Ther; 16(5); 879-92. ©2017 AACR.


Subject(s)
Anthracyclines/administration & dosage , Antibodies, Monoclonal/administration & dosage , Immunoconjugates/administration & dosage , Neoplasms/drug therapy , Ado-Trastuzumab Emtansine , Aminoacyltransferases/chemistry , Animals , Anthracyclines/chemistry , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Bacterial Proteins/chemistry , Brentuximab Vedotin , Cell Line, Tumor , Cysteine Endopeptidases/chemistry , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Doxorubicin/chemistry , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Ki-1 Antigen/chemistry , Ki-1 Antigen/immunology , Maytansine/analogs & derivatives , Maytansine/chemistry , Maytansine/immunology , Mice , Neoplasms/immunology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/immunology , Trastuzumab/administration & dosage , Trastuzumab/chemistry , Trastuzumab/immunology , Xenograft Model Antitumor Assays
6.
Cancer Sci ; 107(7): 1039-46, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27166974

ABSTRACT

Antibody-drug conjugates deliver anticancer agents selectively and efficiently to tumor tissue and have significant antitumor efficacy with a wide therapeutic window. DS-8201a is a human epidermal growth factor receptor 2 (HER2)-targeting antibody-drug conjugate prepared using a novel linker-payload system with a potent topoisomerase I inhibitor, exatecan derivative (DX-8951 derivative, DXd). It was effective against trastuzumab emtansine (T-DM1)-insensitive patient-derived xenograft models with both high and low HER2 expression. In this study, the bystander killing effect of DS-8201a was evaluated and compared with that of T-DM1. We confirmed that the payload of DS-8201a, DXd (1), was highly membrane-permeable whereas that of T-DM1, Lys-SMCC-DM1, had a low level of permeability. Under a coculture condition of HER2-positive KPL-4 cells and negative MDA-MB-468 cells in vitro, DS-8201a killed both cells, whereas T-DM1 and an antibody-drug conjugate with a low permeable payload, anti-HER2-DXd (2), did not. In vivo evaluation was carried out using mice inoculated with a mixture of HER2-positive NCI-N87 cells and HER2-negative MDA-MB-468-Luc cells by using an in vivo imaging system. In vivo, DS-8201a reduced the luciferase signal of the mice, indicating suppression of the MDA-MB-468-Luc population; however, T-DM1 and anti-HER2-DXd (2) did not. Furthermore, it was confirmed that DS-8201a was not effective against MDA-MB-468-Luc tumors inoculated at the opposite side of the NCI-N87 tumor, suggesting that the bystander killing effect of DS-8201a is observed only in cells neighboring HER2-positive cells, indicating low concern in terms of systemic toxicity. These results indicated that DS-8201a has a potent bystander effect due to a highly membrane-permeable payload and is beneficial in treating tumors with HER2 heterogeneity that are unresponsive to T-DM1.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Bystander Effect/drug effects , Camptothecin/analogs & derivatives , Immunoconjugates/immunology , Immunoconjugates/pharmacology , Neoplasms/genetics , Neoplasms/pathology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Ado-Trastuzumab Emtansine , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Camptothecin/immunology , Camptothecin/pharmacology , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Cell Proliferation/drug effects , Female , Humans , Maytansine/analogs & derivatives , Maytansine/immunology , Maytansine/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/enzymology , Neoplasms/immunology , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Topoisomerase I Inhibitors/pharmacology , Trastuzumab
7.
MAbs ; 7(6): 989-1009, 2015.
Article in English | MEDLINE | ID: mdl-26292154

ABSTRACT

Antibody-drug conjugates are highly complex entities that combine an antibody, a linker and a toxin. This complexity makes them demanding both technically and from a regulatory point of view, and difficult to deal with in their patent aspects. This article discusses different issues of patent protection and freedom to operate with regard to this promising new class of drugs.


Subject(s)
Antibodies, Monoclonal/immunology , Drug Approval , Immunoconjugates/immunology , Intellectual Property , Ado-Trastuzumab Emtansine , Aminoglycosides/immunology , Aminoglycosides/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Brentuximab Vedotin , Gemtuzumab , Humans , Immunoconjugates/therapeutic use , Maytansine/analogs & derivatives , Maytansine/immunology , Maytansine/therapeutic use , Patents as Topic , Trastuzumab , United States , United States Food and Drug Administration
8.
PLoS One ; 10(7): e0131177, 2015.
Article in English | MEDLINE | ID: mdl-26132162

ABSTRACT

Antibody drug conjugates (ADCs) have recently been proven to be highly potent anti-tumor drugs, typically exceeding the efficacy of conventional monoclonal antibodies (mAbs). ADCs are currently produced by chemical conjugation of a small-molecule toxin to the mAb through lysine or cysteine side chains. This leads to heterogeneous mixtures of ADCs in which variable numbers of drugs are conjugated to individual antibodies and in which the site of conjugation cannot be defined. Consequently, there is currently significant interest in further development of drug conjugation technologies, with a particular focus on site-specific payload conjugation. Here, we present an enzymatic conjugation platform based on the S. aureus sortase A-mediated transpeptidation reaction, allowing the efficient generation of ADCs with toxins conjugated to pre-defined sites at pre-defined drug-to-antibody ratios. For this, two modifications were introduced: first, immunoglobulin heavy (IgH) and light (IgL) chains were modified at their C-termini by addition of the sortase A recognition motif LPETG, and second, the small molecule tubulin polymerization inhibitors monomethylauristatin E (MMAE) and maytansine were modified by addition of a pentaglycine peptide, thus making them suitable substrates for sortase A-mediated transpeptidation. We demonstrate efficient generation and characterization of the anti-CD30 ADC Ac10-vcPAB-MMAE, an enzymatically conjugated counterpart of brentuximab vedotin (Adcetris), as well as several anti-HER-2 ADCs including trastuzumab-maytansine, the counterpart of trastuzumab emtansine (Kadcyla). ADCs generated in this manner were found to display in vitro cell killing activities indistinguishable from the classic conjugates. Further, when tested in vivo in a HER-2-overexpressing ovarian cancer xenograft mouse model, enzymatically generated trastuzumab-maytansine was found to lead to complete regression of established tumors, similar to Kadcyla.


Subject(s)
Aminoacyltransferases/chemistry , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Bacterial Proteins/chemistry , Cysteine Endopeptidases/chemistry , Immunoconjugates/pharmacology , Maytansine/analogs & derivatives , Ovarian Neoplasms/drug therapy , Ado-Trastuzumab Emtansine , Aminoacyltransferases/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Bacterial Proteins/immunology , Brentuximab Vedotin , Cysteine Endopeptidases/immunology , Female , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Ki-1 Antigen/antagonists & inhibitors , Ki-1 Antigen/genetics , Ki-1 Antigen/immunology , Maytansine/chemistry , Maytansine/immunology , Maytansine/pharmacology , Mice , Mice, Nude , Oligopeptides/chemistry , Oligopeptides/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Protein Engineering , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Staphylococcus aureus/chemistry , Staphylococcus aureus/enzymology , Trastuzumab , Xenograft Model Antitumor Assays
9.
Mol Cancer Ther ; 14(7): 1614-24, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25931519

ABSTRACT

Epidermal growth factor receptor variant III (EGFRvIII) is a cancer-specific deletion mutant observed in approximately 25% to 50% of glioblastoma multiforme (GBM) patients. An antibody drug conjugate, AMG 595, composed of the maytansinoid DM1 attached to a highly selective anti-EGFRvIII antibody via a noncleavable linker, was developed to treat EGFRvIII-positive GBM patients. AMG 595 binds to the cell surface and internalizes into the endo-lysosomal pathway of EGFRvIII-expressing cells. Incubation of AMG 595 with U251 cells expressing EGFRvIII led to potent growth inhibition. AMG 595 treatment induced significant tumor mitotic arrest, as measured by phospho-histone H3, in GBM subcutaneous xenografts expressing EGFRvIII. A single intravenous injection of AMG 595 at 17 mg/kg (250 µg DM1/kg) generated complete tumor regression in the U251vIII subcutaneous xenograft model. AMG 595 mediated tumor regression in the D317 subcutaneous xenograft model that endogenously expresses EGFRvIII. Finally, AMG 595 treatment inhibited the growth of D317 xenografts orthotopically implanted into the brain as determined by magnetic resonance imaging. These results demonstrate that AMG 595 is a promising candidate to evaluate in EGFRvIII-expressing GBM patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , Brain Neoplasms/drug therapy , ErbB Receptors/immunology , Glioblastoma/drug therapy , Immunoconjugates/pharmacology , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Glioblastoma/immunology , Glioblastoma/metabolism , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/immunology , Immunohistochemistry , Injections, Intravenous , Maytansine/analogs & derivatives , Maytansine/immunology , Maytansine/pharmacology , Mice, Nude , Mice, SCID , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/immunology
10.
Mol Cancer Ther ; 14(7): 1605-13, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25904506

ABSTRACT

A majority of ovarian and non-small cell lung adenocarcinoma cancers overexpress folate receptor α (FRα). Here, we report the development of an anti-FRα antibody-drug conjugate (ADC), consisting of a FRα-binding antibody attached to a highly potent maytansinoid that induces cell-cycle arrest and cell death by targeting microtubules. From screening a large panel of anti-FRα monoclonal antibodies, we selected the humanized antibody M9346A as the best antibody for targeted delivery of a maytansinoid payload into FRα-positive cells. We compared M9346A conjugates with various linker/maytansinoid combinations, and found that a conjugate, now denoted as IMGN853, with the N-succinimidyl 4-(2-pyridyldithio)-2-sulfobutanoate (sulfo-SPDB) linker and N(2')-deacetyl-N(2')-(4-mercapto-4-methyl-1-oxopentyl)-maytansine (DM4) exhibited the most potent antitumor activity in several FRα-expressing xenograft tumor models. The level of expression of FRα on the surface of cells was a major determinant in the sensitivity of tumor cells to the cytotoxic effect of the conjugate. Efficacy studies of IMGN853 in xenografts of ovarian cancer and non-small cell lung cancer cell lines and of a patient tumor-derived xenograft model demonstrated that the ADC was highly active against tumors that expressed FRα at levels similar to those found on a large fraction of ovarian and non-small cell lung cancer patient tumors, as assessed by immunohistochemistry. IMGN853 displayed cytotoxic activity against FRα-negative cells situated near FRα-positive cells (bystander cytotoxic activity), indicating its ability to eradicate tumors with heterogeneous expression of FRα. Together, these findings support the clinical development of IMGN853 as a novel targeted therapy for patients with FRα-expressing tumors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Folate Receptor 1/antagonists & inhibitors , Immunoconjugates/pharmacology , Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Bystander Effect/drug effects , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Female , Folate Receptor 1/immunology , Humans , Immunoconjugates/immunology , Maytansine/analogs & derivatives , Maytansine/immunology , Maytansine/pharmacology , Mice, Nude , Mice, SCID , Molecular Targeted Therapy/methods , Neoplasms/immunology , Neoplasms/metabolism , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/immunology
12.
MAbs ; 6(2): 556-66, 2014.
Article in English | MEDLINE | ID: mdl-24492307

ABSTRACT

Lorvotuzumab mertansine (LM) is an antibody-drug conjugate composed of a humanized anti-CD56 antibody, lorvotuzumab, linked via a cleavable disulfide linker to the tubulin-binding maytansinoid DM1. CD56 is expressed on most small cell lung cancers (SCLC), providing a promising therapeutic target for treatment of this aggressive cancer, which has a poor five-year survival rate of only 5-10%. We performed immunohistochemical staining on SCLC tumor microarrays, which confirmed that CD56 is expressed at high levels on most (~74%) SCLC tumors. Conjugation of lorvotuzumab with DM1 did not alter its specific binding to cells and LM demonstrated potent target-dependent cytotoxicity against CD56-positive SCLC cells in vitro. The anti-tumor activity of LM was evaluated against SCLC xenograft models in mice, both as monotherapy and in combination with platinum/etoposide and paclitaxel/carboplatin. Dose-dependent and antigen-specific anti-tumor activity of LM monotherapy was demonstrated at doses as low as 3 mg/kg. LM was highly active in combination with standard-of-care platinum/etoposide therapies, even in relatively resistant xenograft models. LM demonstrated outstanding anti-tumor activity in combination with carboplatin/etoposide, with superior activity over chemotherapy alone when LM was used in combinations at significantly reduced doses (6-fold below the minimally efficacious dose for LM monotherapy). The combination of LM with carboplatin/paclitaxel was also highly active. This study provides the rationale for clinical evaluation of LM as a promising novel targeted therapy for SCLC, both as monotherapy and in combination with chemotherapy.


Subject(s)
Antibodies, Monoclonal/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD56 Antigen/immunology , Immunotherapy/methods , Lung Neoplasms/therapy , Maytansine/analogs & derivatives , Maytansine/metabolism , Small Cell Lung Carcinoma/therapy , Tubulin Modulators/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Drug Synergism , Drug Therapy, Combination , Female , Humans , Lung Neoplasms/immunology , Maytansine/chemistry , Maytansine/immunology , Mice , Mice, SCID , Small Cell Lung Carcinoma/immunology , Tubulin Modulators/chemistry , Xenograft Model Antitumor Assays
13.
Bioanalysis ; 5(9): 1007-23, 2013 May.
Article in English | MEDLINE | ID: mdl-23641693

ABSTRACT

BACKGROUND: Antibody-drug conjugates (ADCs) such as Kadcyla™ (ado-trastuzumab emtansine [T-DM1]) present covalently bound cytotoxic drugs, which may influence their immunogenicity potential compared with antibody therapies. Therefore, ADCs require assay strategies that allow measurement of responses to all the molecular components. RESULTS: The immunogenicity strategy for T-DM1 used a risk-based, tiered approach that included screening and titration to detect antitherapeutic antibodies; confirmation of positive responses; and characterization to assess whether the immune response is primarily to the antibody or to the linker-drug and/or new epitopes in trastuzumab resulting from conjugation. CONCLUSION: The tiered immunogenicity assay strategy for T-DM1 allowed detection of antitherapeutic antibodies to all components of the ADC in multiple nonclinical and clinical studies. Characterization strategies implemented in clinical studies provided additional insights into the specificity of the immune response.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal/immunology , Immunogenetic Phenomena , Maytansine/analogs & derivatives , Ado-Trastuzumab Emtansine , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Humans , Macaca fascicularis , Maytansine/administration & dosage , Maytansine/immunology , Maytansine/therapeutic use , Trastuzumab
14.
MAbs ; 5(1): 5-12, 2013.
Article in English | MEDLINE | ID: mdl-23255090

ABSTRACT

The American Association of Pharmaceutical Scientists (AAPS) National Biotechnology Conference Short Course "Translational Challenges in Developing Antibody-Drug Conjugates (ADCs)," held May 24, 2012 in San Diego, CA, was organized by members of the Pharmacokinetics, Pharmacodynamics and Drug Metabolism section of AAPS. Representatives from the pharmaceutical industry, regulatory authorities, and academia in the US and Europe attended this short course to discuss the translational challenges in ADC development and the importance of characterizing these molecules early in development to achieve therapeutic utility in patients. Other areas of discussion included selection of target antigens; characterization of absorption, distribution, metabolism, and excretion; assay development and hot topics like regulatory perspectives and the role of pharmacometrics in ADC development. MUC16-targeted ADCs were discussed to illustrate challenges in preclinical development; experiences with trastuzumab emtansine (T-DM1; Genentech) and the recently approved brentuximab vedotin (Adcetris; Seattle Genetics) were presented in depth to demonstrate considerations in clinical development. The views expressed in this report are those of the participants and do not necessarily represent those of their affiliations.


Subject(s)
Antibodies, Monoclonal , Antineoplastic Agents , Immunoconjugates , Neoplasms/therapy , Translational Research, Biomedical/trends , Ado-Trastuzumab Emtansine , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Biotechnology/methods , Brentuximab Vedotin , Clinical Trials as Topic , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Maytansine/analogs & derivatives , Maytansine/immunology , Maytansine/pharmacokinetics , Maytansine/therapeutic use , Neoplasms/immunology , Translational Research, Biomedical/methods , Trastuzumab
15.
Cancer ; 118(23): 5733-40, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22648179

ABSTRACT

BACKGROUND: We conducted a phase 1, multicenter, open-label, dose-escalation study (TDM3569g) to assess the safety, tolerability, and pharmacokinetics of single-agent trastuzumab emtansine (T-DM1) administered weekly and once every 3 weeks in patients with HER2-positive metastatic breast cancer previously treated with trastuzumab. The weekly dose results are described here. METHODS: Patients were administered escalating doses of T-DM1 weekly, starting at 1.2 mg/kg. Additional patients were enrolled at the maximum tolerated dose (MTD) to better characterize tolerability and pharmacokinetics. RESULTS: Twenty-eight patients received weekly T-DM1, and the MTD was determined to be 2.4 mg/kg. In general, T-DM1 was well tolerated, requiring few dose modifications or discontinuations because of adverse events (AEs). Grade ≥ 3 AEs were reported in 19 patients (67.9%); treatment-related AEs occurred in 25 (89.3%) patients. Exposure to weekly T-DM1 was dose-proportional at ≥ 1.2 mg/kg, and accumulation of T-DM1 and total trastuzumab was observed. Objective partial tumor responses were reported in 13 (46.4%) patients; the median duration of response was 18.6 months, and the 6-month clinical benefit rate was 57.1%. CONCLUSION: The results suggest that a weekly dose of T-DM1 2.4 mg/kg has antitumor activity and is well tolerated in patients with HER2-positive metastatic breast cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Maytansine/analogs & derivatives , Receptor, ErbB-2/analysis , Ado-Trastuzumab Emtansine , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Agents/adverse effects , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/chemistry , Female , Humans , Immunotoxins/administration & dosage , Immunotoxins/adverse effects , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Maximum Tolerated Dose , Maytansine/administration & dosage , Maytansine/adverse effects , Maytansine/immunology , Maytansine/pharmacokinetics , Middle Aged , Trastuzumab
16.
Nat Biotechnol ; 30(2): 184-9, 2012 Jan 22.
Article in English | MEDLINE | ID: mdl-22267010

ABSTRACT

The reactive thiol in cysteine is used for coupling maleimide linkers in the generation of antibody conjugates. To assess the impact of the conjugation site, we engineered cysteines into a therapeutic HER2/neu antibody at three sites differing in solvent accessibility and local charge. The highly solvent-accessible site rapidly lost conjugated thiol-reactive linkers in plasma owing to maleimide exchange with reactive thiols in albumin, free cysteine or glutathione. In contrast, a partially accessible site with a positively charged environment promoted hydrolysis of the succinimide ring in the linker, thereby preventing this exchange reaction. The site with partial solvent-accessibility and neutral charge displayed both properties. In a mouse mammary tumor model, the stability and therapeutic activity of the antibody conjugate were affected positively by succinimide ring hydrolysis and negatively by maleimide exchange with thiol-reactive constituents in plasma. Thus, the chemical and structural dynamics of the conjugation site can influence antibody conjugate performance by modulating the stability of the antibody-linker interface.


Subject(s)
Antibodies/blood , Antibodies/immunology , Binding Sites, Antibody/immunology , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunoglobulin G/chemistry , Protein Engineering , Aminobenzoates/chemistry , Aminobenzoates/immunology , Animals , Antibodies/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Cell Line, Tumor , Cell Survival , Cysteine/chemistry , Humans , Immunoconjugates/administration & dosage , Immunoglobulin G/immunology , Macaca fascicularis , Maleimides/chemistry , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Maytansine/chemistry , Maytansine/immunology , Mice , Mice, Nude , Models, Molecular , Oligopeptides/chemistry , Oligopeptides/immunology , Protein Conformation , Rats , Structure-Activity Relationship , Sulfhydryl Compounds/chemistry , Trastuzumab
17.
Int J Mol Sci ; 13(12): 16020-45, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23443108

ABSTRACT

Drug conjugates have been studied extensively in preclinical in vitro and in vivo models but to date only a few compounds have progressed to the clinical setting. This situation is now changing with the publication of studies demonstrating a significant impact on clinical practice and highlighting the potential of this new class of targeted therapies. This review summarizes the pharmacological and molecular background of the main drug conjugation systems, namely antibody drug conjugates (ADCs), immunotoxins and immunoliposomes. All these compounds combine the specific targeting moiety of an antibody or similar construct with the efficacy of a toxic drug. The aim of this strategy is to target tumor cells specifically while sparing normal tissue, thus resulting in high efficacy and low toxicity. Recently, several strategies have been investigated in phase I clinical trials and some have entered phase III clinical development. This review provides a detailed overview of various strategies and critically discusses the most relevant achievements. Examples of the most advanced compounds include T-DM1 and brentuximab vedotin. However, additional promising strategies such as immunotoxins and immunoliposmes are already in clinical development. In summary, targeted drug delivery by drug conjugates is a new emerging class of anti-cancer therapy that may play a major role in the future.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Agents , Immunoconjugates , Immunotoxins , Liposomes , Maytansine/analogs & derivatives , Neoplasms , Ado-Trastuzumab Emtansine , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Brentuximab Vedotin , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Immunotoxins/therapeutic use , Liposomes/immunology , Liposomes/pharmacokinetics , Liposomes/therapeutic use , Maytansine/immunology , Maytansine/pharmacokinetics , Maytansine/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Trastuzumab
18.
Curr Opin Oncol ; 23(6): 594-600, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21986845

ABSTRACT

PURPOSE OF REVIEW: In this review, we aim to update the clinical data of trastuzumab-DM1 (T-DM1) in terms of safety and efficacy, and describe ongoing and future trials evaluating its potential role in the management of patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancer. RECENT FINDINGS: Trastuzumab emtansine (T-DM1) is an antibody drug conjugate that optimizes delivery of chemotherapy with an anti-HER2 monoclonal antibody. As a conjugate, T-DM1's systemic side effects are significantly minimized due to its targeted delivery by trastuzumab to HER2-positive cells. Phase I and II studies show that the maximum tolerated dose, and thus the recommended dose for T-DM1, is 3.6  mg/kg given intravenously every 3 weeks. Single arm phase Ib/II, II and a randomized phase II first-line study of T-DM1 versus the combination of trastuzumab + docetaxel all showed improved tolerability, and at least equivalent efficacy, compared with our current standard of care. Two randomized phase III registration studies are now active, evaluating this agent in the refractory and first-line HER2-positive settings. SUMMARY: T-DM1 has been shown to be a very promising agent for the targeted delivery of chemotherapy and anti-HER2 monoclonal antibody therapy for patients with metastatic, HER2-positive breast cancer. T-DM1 will likely play a role in the management of patients with advanced and early stage HER2-positive breast cancer, but this awaits further study.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Breast Neoplasms/enzymology , Breast Neoplasms/therapy , Immunotoxins/therapeutic use , Maytansine/analogs & derivatives , Receptor, ErbB-2/antagonists & inhibitors , Ado-Trastuzumab Emtansine , Antibodies, Monoclonal, Humanized/immunology , Clinical Trials, Phase III as Topic , Female , Humans , Immunotoxins/immunology , Maytansine/immunology , Maytansine/therapeutic use , Randomized Controlled Trials as Topic , Receptor, ErbB-2/immunology , Trastuzumab
19.
MAbs ; 3(4): 331-7, 2011.
Article in English | MEDLINE | ID: mdl-21691144

ABSTRACT

The World Antibody Drug Conjugate Summit Europe, organized by Biorbis/ Hanson Wade was held in Frankfurt, Germany on February 21-23, 2011. Antibody drug conjugates (ADCs), also called immunoconjugates, are becoming an increasingly important class of therapeutics as demonstrated by the attendance of nearly 100 delegates at this highly focused meeting. Updates on three ADCs that are in late-stage clinical development, trastuzumab emtansine (T-DM1), brentuximab vedotin (SGN-35) and inotuzumab ozogamicin (CMC-544), were presented by speakers from ImmunoGen, Genentech, Roche, Seattle Genetics and Pfizer. These ADCs have shown encouraging therapeutic effects against solid tumors (T-DM1) and hematological malignancies (SGN-35, CMC-544). The key feature of the new generation of ADCs is the effective combination of the cytotoxicity of natural or synthetic highly potent antineoplastic agents, tumor selective monoclonal antibodies and blood-stable optimized linkers. Early clinical data for ADCs were showcased by Progenics Pharmaceuticals (PSMA ADC), Celldex (CDX-011) and Biotest (BT-062). Takeda, MedImmune and sanofi-aventis outlined their strategies for process development and analytical characterization. In addition, presentations on duocarmycin based-ADCs, alpha emitting immunoconjugates and antibody-conjugated nanoparticles were given by representatives from Syntarga, Algeta and the University of Stuttgart, respectively.


Subject(s)
Antibodies, Monoclonal , Antineoplastic Agents/therapeutic use , Immunoconjugates , Immunotherapy/trends , Neoplasms/therapy , Ado-Trastuzumab Emtansine , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Biotechnology/trends , Brentuximab Vedotin , Clinical Trials as Topic , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunoconjugates/therapeutic use , Inotuzumab Ozogamicin , Maytansine/analogs & derivatives , Maytansine/chemistry , Maytansine/immunology , Maytansine/therapeutic use , Trastuzumab
20.
Mol Cancer Ther ; 9(10): 2689-99, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20937594

ABSTRACT

Maytansine is a potent microtubule-targeted compound that induces mitotic arrest and kills tumor cells at subnanomolar concentrations. However, its side effects and lack of tumor specificity have prevented successful clinical use. Recently, antibody-conjugated maytansine derivatives have been developed to overcome these drawbacks. Several conjugates show promising early clinical results. We evaluated the effects on microtubule polymerization and dynamic instability of maytansine and two cellular metabolites (S-methyl-DM1 and S-methyl-DM4) of antibody-maytansinoid conjugates that are potent in cells at picomolar levels and that are active in tumor-bearing mice. Although S-methyl-DM1 and S-methyl-DM4 inhibited polymerization more weakly than maytansine, at 100 nmol/L they suppressed dynamic instability more strongly than maytansine (by 84% and 73%, respectively, compared with 45% for maytansine). However, unlike maytansine, S-methyl-DM1 and S-methyl-DM4 induced tubulin aggregates detectable by electron microscopy at concentrations ≥2 µmol/L, with S-methyl-DM4 showing more extensive aggregate formation than S-methyl-DM1. Both maytansine and S-methyl-DM1 bound to tubulin with similar K(D) values (0.86 ± 0.2 and 0.93 ± 0.2 µmol/L, respectively). Tritiated S-methyl-DM1 bound to 37 high-affinity sites per microtubule (K(D), 0.1 ± 0.05 µmol/L). Thus, S-methyl-DM1 binds to high-affinity sites on microtubules 20-fold more strongly than vinblastine. The high-affinity binding is likely at microtubule ends and is responsible for suppression of microtubule dynamic instability. Also, at higher concentrations, S-methyl-DM1 showed low-affinity binding either to a larger number of sites on microtubules or to sedimentable tubulin aggregates. Overall, the maytansine derivatives that result from cellular metabolism of the antibody conjugates are themselves potent microtubule poisons, interacting with microtubules as effectively as or more effectively than the parent molecule.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Immunoconjugates/immunology , Maytansine/pharmacology , Microtubules/drug effects , Animals , Antineoplastic Agents, Phytogenic/immunology , Antineoplastic Agents, Phytogenic/metabolism , Binding Sites , Cattle , Maytansine/immunology , Maytansine/metabolism , Mice , Microtubules/metabolism , Polymers , Solubility , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...