Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 20(1): 203-212, 2021 01.
Article in English | MEDLINE | ID: mdl-33177153

ABSTRACT

Several antibody-drug conjugates (ADC) showing strong clinical responses in solid tumors target high expression antigens (HER2, TROP2, Nectin-4, and folate receptor alpha/FRα). Highly expressed tumor antigens often have significant low-level expression in normal tissues, resulting in the potential for target-mediated drug disposition (TMDD) and increased clearance. However, ADCs often do not cross-react with normal tissue in animal models used to test efficacy (typically mice), and the impact of ADC binding to normal tissue antigens on tumor response remains unclear. An antibody that cross-reacts with human and murine FRα was generated and tested in an animal model where the antibody/ADC bind both human tumor FRα and mouse FRα in normal tissue. Previous work has demonstrated that a "carrier" dose of unconjugated antibody can improve the tumor penetration of ADCs with high expression target-antigens. A carrier dose was employed to study the impact on cross-reactive ADC clearance, distribution, and efficacy. Co-administration of unconjugated anti-FRα antibody with the ADC-improved efficacy, even in low expression models where co-administration normally lowers efficacy. By reducing target-antigen-mediated clearance in normal tissue, the co-administered antibody increased systemic exposure, improved tumor tissue penetration, reduced target-antigen-mediated uptake in normal tissue, and increased ADC efficacy. However, payload potency and tumor antigen saturation are also critical to efficacy, as shown with reduced efficacy using too high of a carrier dose. The judicious use of higher antibody doses, either through lower DAR or carrier doses, can improve the therapeutic window by increasing efficacy while lowering target-mediated toxicity in normal tissue.


Subject(s)
Antibodies/administration & dosage , Antibodies/pharmacology , Immunoconjugates/metabolism , Animals , Antibodies/toxicity , Cell Line, Tumor , Cross Reactions/immunology , Drug Carriers/chemistry , Female , Immunoconjugates/blood , Mice , Mice, SCID , Neoplasms/pathology , Treatment Outcome
2.
ACS Med Chem Lett ; 10(10): 1393-1399, 2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31620224

ABSTRACT

A new type of antibody-drug conjugate (ADC) has been prepared that contains a sulfur-bearing maytansinoid attached to an antibody via a highly stable tripeptide linker. Once internalized by cells, proteases in catabolic vesicles cleave the peptide of the ADC's linker causing self-immolation that releases a thiol-bearing metabolite, which is then S-methylated. Conjugates were prepared with peptide linkers containing only alanyl residues, which were all l isomers or had a single d residue in one of the three positions. A d-alanyl residue in the linker did not significantly impair a conjugate's cytotoxicity or bystander killing unless it was directly attached to the immolative moiety. Increasing the number of methylene units in the maytansinoid side chain of a conjugate did not typically affect an ADC's cytotoxicity to targeted cells but did increase bystander killing activity. ADCs with the highest in vitro bystander killing were then evaluated in vivo in mice, where they displayed improved efficacy compared to previously described types of maytansinoid conjugates.

3.
ACS Med Chem Lett ; 10(8): 1211-1215, 2019 Aug 08.
Article in English | MEDLINE | ID: mdl-31413807

ABSTRACT

Indolinobenzodiazepine DNA alkylators (IGNs) are the cytotoxic payloads in antibody-drug conjugates (ADCs) currently undergoing Phase I clinical evaluation (IMGN779, IMGN632, and TAK164). These ADCs possess linkers that have been incorporated into a central substituted phenyl spacer. Here, we present an alternative strategy for the IGNs, linking through a carbamate at the readily available N-10 amine present in the monoimine containing dimer. As a result, we have designed a series of N-10 linked IGN ADCs with a wide range of in vitro potency and tolerability, which may allow us to better match an IGN with a particular target based on the potential dosing needs.

4.
Bioorg Med Chem Lett ; 29(17): 2455-2458, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31350125

ABSTRACT

Antibody-drug conjugates (ADCs) incorporating potent indolinobenzodiazepine (IGN) DNA alkylators as the cytotoxic payload are currently undergoing clinical evaluation. The optimized design of these payloads consists of an unsymmetrical dimer possessing both an imine and an amine effectively eliminating DNA crosslinking and demonstrating improved tolerability in mice. Here we present an alternate approach to generating DNA alkylating ADCs by linking the IGN monomer with a biaryl system which has a high DNA binding affinity to potentially enhance tolerability. These BIA ADCs were found to be highly cytotoxic in vitro and demonstrated potent antitumor activity in vivo.


Subject(s)
Alkylating Agents/chemistry , Drug Design , Immunoconjugates/chemistry , Animals , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Cell Survival/drug effects , DNA/metabolism , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Mice , Mice, SCID , Neoplasms/drug therapy , Neoplasms/pathology , Structure-Activity Relationship , Transplantation, Heterologous
5.
EJNMMI Res ; 8(1): 87, 2018 Aug 28.
Article in English | MEDLINE | ID: mdl-30155674

ABSTRACT

BACKGROUND: The folate receptor α (FRα)-targeting antibody-drug conjugate (ADC), IMGN853, shows great antitumor activity against FRα-expressing tumors in vivo, but patient selection and consequently therapy outcome are based on immunohistochemistry. The aim of this study is to develop an antibody-derived immuno-PET imaging agent strategy for targeting FRα in ovarian cancer as a predictor of treatment success. METHODS: We developed [89Zr]Zr-DFO-M9346A, a humanized antibody-based radiotracer targeting tumor-associated FRα in the preclinical setting. [89Zr]Zr-DFO-M9346A's binding ability was tested in an in vitro uptake assay using cell lines with varying FRα expression levels. The diagnostic potential of [89Zr]Zr-M9346A was evaluated in KB and OV90 subcutaneous xenografts. Following intravenous injection of [89Zr]Zr-DFO-M9346A (~90 µCi, 50 µg), PET imaging and biodistribution studies were performed. We determined the blood half-life of [89Zr]Zr-DFO-M9346A and compared it to the therapeutic, radioiodinated ADC [131I]-IMGN853. Finally, in vivo studies using IMG853 as a therapeutic, paired with [89Zr]Zr-DFO-M9346A as a companion diagnostic were performed using OV90 xenografts. RESULTS: DFO-M9346A was labeled with Zr-89 at 37 °C within 60 min and isolated in labeling yields of 85.7 ± 5.7%, radiochemical purities of 98.0 ± 0.7%, and specific activities of 3.08 ± 0.43 mCi/mg. We observed high specificity for binding FRα positive cells in vitro. For PET and biodistribution studies, [89Zr]Zr-M9346A displayed remarkable in vivo performance in terms of excellent tumor uptake for KB and OV xenografts (45.8 ± 29.0 %IA/g and 26.1 ± 7.2 %IA/g), with low non-target tissue uptake in other organs such as kidneys (4.5 ± 1.2 %IA/g and 4.3 ± 0.7 %IA/g). A direct comparison of the blood half life of [89Zr]Zr-M9346A and [131I]-IMGN853 corroborated the equivalency of the radiopharmaceutical and the ADC, paving the way for a companion PET imaging study. CONCLUSIONS: We developed a new folate receptor-targeted 89Zr-labeled PET imaging agent with excellent pharmacokinetics in vivo. Good tumor uptake in subcutaneous KB and OV90 xenografts were obtained, and ADC therapy studies were performed with the precision predictor.

6.
Mol Cancer Ther ; 17(3): 650-660, 2018 03.
Article in English | MEDLINE | ID: mdl-29440292

ABSTRACT

Tumor-selective delivery of cytotoxic agents in the form of antibody-drug conjugates (ADCs) is now a clinically validated approach for cancer treatment. In an attempt to improve the clinical success rate of ADCs, emphasis has been recently placed on the use of DNA-cross-linking pyrrolobenzodiazepine compounds as the payload. Despite promising early clinical results with this class of ADCs, doses achievable have been low due to systemic toxicity. Here, we describe the development of a new class of potent DNA-interacting agents wherein changing the mechanism of action from a cross-linker to a DNA alkylator improves the tolerability of the ADC. ADCs containing the DNA alkylator displayed similar in vitro potency, but improved bystander killing and in vivo efficacy, compared with those of the cross-linker. Thus, the improved in vivo tolerability and antitumor activity achieved in rodent models with ADCs of the novel DNA alkylator could provide an efficacious, yet safer option for cancer treatment. Mol Cancer Ther; 17(3); 650-60. ©2018 AACR.


Subject(s)
Immunoconjugates/pharmacology , Intercalating Agents/pharmacology , Neoplasms/drug therapy , Therapeutic Index, Drug , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cross-Linking Reagents/chemistry , DNA/genetics , DNA/metabolism , Drug Design , Humans , Immunoconjugates/chemistry , Immunoconjugates/metabolism , Intercalating Agents/chemistry , Intercalating Agents/metabolism , Mice , Neoplasms/pathology , Tumor Burden/drug effects
7.
Mol Cancer Ther ; 17(5): 1003-1011, 2018 05.
Article in English | MEDLINE | ID: mdl-29440294

ABSTRACT

Grade 3 endometrioid and uterine serous carcinomas (USC) account for the vast majority of endometrial cancer deaths. The purpose of this study was to determine folic acid receptor alpha (FRα) expression in these biologically aggressive (type II) endometrial cancers and evaluate FRα as a targetable receptor for IMGN853 (mirvetuximab soravtansine). The expression of FRα was evaluated by immunohistochemistry (IHC) and flow cytometry in 90 endometrioid and USC samples. The in vitro cytotoxic activity and bystander effect were studied in primary uterine cancer cell lines expressing differential levels of FRα. In vivo antitumor efficacy of IMGN853 was evaluated in xenograft/patient-derived xenograft (PDX) models. Semiquantitative IHC analysis indicated that 41% of the USC patients overexpress FRα. Further, overexpression of FRα (i.e., 2+) was detected via flow cytometry in 22% (2/9) of primary endometrioid and in 27% (3/11) of primary USC cell lines. Increased cytotoxicity was seen with IMGN853 treatment compared with control in 2+ expressing uterine tumor cell lines. In contrast, tumor cell lines with low FRα showed no difference when exposed to IMGN853 versus control. IMGN853 induced bystander killing of FRα = 0 tumor cells. In an endometrioid xenograft model (END(K)265), harboring 2+ FRα, IMGN853 treatment showed complete resolution of tumors (P < 0.001). Treatment with IMGN853 in the USC PDX model (BIO(K)1), expressing 2+ FRα, induced twofold increase in median survival (P < 0.001). IMGN853 shows impressive antitumor activity in biologically aggressive FRα 2+ uterine cancers. These preclinical data suggest that patients with chemotherapy resistant/recurrent endometrial cancer overexpressing FRα may benefit from this treatment. Mol Cancer Ther; 17(5); 1003-11. ©2018 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Endometrial Neoplasms/drug therapy , Folate Receptor 1/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/chemistry , Antineoplastic Combined Chemotherapy Protocols/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Folate Receptor 1/metabolism , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Maytansine/administration & dosage , Maytansine/analogs & derivatives , Maytansine/chemistry , Mice, SCID , Retrospective Studies
8.
Future Oncol ; 14(2): 123-136, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29098867

ABSTRACT

Resistance to platinum-based therapy poses a significant clinical challenge for the management of advanced ovarian cancer, a leading cause of cancer mortality among women. Mirvetuximab soravtansine is a novel antibody-drug conjugate that targets folate receptor-α, a validated molecular target for therapeutic intervention in this disease. Here, we examine mirvetuximab soravtansine's mechanism of action and pharmacology, and review its clinical evaluation in ovarian cancer to date. We focus on the favorable tolerability and encouraging signals of efficacy that have emerged, most notably in patients with platinum-resistant disease. Ongoing Phase III monotherapy and Phase Ib/II combination trials evaluating its activity in the setting of platinum resistance are emphasized, which will help define its role in the evolving landscape of ovarian cancer therapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Drug Resistance, Neoplasm/genetics , Immunoconjugates/therapeutic use , Maytansine/analogs & derivatives , Ovarian Neoplasms/drug therapy , Female , Folate Receptor 1/antagonists & inhibitors , Folate Receptor 1/immunology , Humans , Immunoconjugates/immunology , Maytansine/therapeutic use , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Platinum/adverse effects , Platinum/therapeutic use
9.
Neoplasia ; 19(9): 661-671, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28753442

ABSTRACT

Naratuximab emtansine (IMGN529) is an investigational antibody-drug conjugate consisting of a CD37-targeting antibody conjugated to the maytansine-derived microtuble disruptor, DM1. IMGN529 has shown promising preclinical and clinical activity in non-Hodgkin lymphoma, including diffuse large B-cell lymphoma (DLBCL). Due to the aggressive nature of the disease, DLBCL is often treated with combination therapies to maximize clinical outcomes; therefore, we investigated the potential of combining IMGN529 with both standard-of-care and emerging therapies against multiple oncology-relevant targets and pathways. The strongest enhancement in potency was seen with anti-CD20 antibodies, including rituximab. The combination of IMGN529 and rituximab was more potent than either agent alone, and this combinatorial benefit was associated with increased apoptotic induction and cell death. Additional studies revealed that rituximab treatment increased the internalization and degradation of the CD37-targeting antibody moiety of IMGN529. The combination of IMGN529 and rituximab was highly efficacious in multiple xenograft models, with superior antitumor efficacy seen compared to either agent alone or treatment with R-CHOP therapy. These findings suggest a novel mechanism whereby the potency of IMGN529 can be enhanced by CD20 binding, which results in the increased internalization and degradation of IMGN529 leading to the generation of greater amounts of cytotoxic catabolite. Overall, these data provide a biological rationale for the enhanced activity of IMGN529 in combination with rituximab and support the ongoing clinical evaluation of IMGN529 in combination with rituximab in patients with relapsed and/or refractory DLBCL.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Lymphoma, Non-Hodgkin/drug therapy , Rituximab/pharmacology , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Female , Humans , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/pathology , Mice , Molecular Targeted Therapy , Proteolysis , Xenograft Model Antitumor Assays
10.
Cancer ; 123(16): 3080-3087, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28440955

ABSTRACT

BACKGROUND: Mirvetuximab soravtansine (IMGN853) is an antibody-drug conjugate that selectively targets folate receptor α (FRα). In this phase 1 dose-escalation study, the authors investigated IMGN853 in patients with FRα-positive solid tumors. METHODS: Patients received IMGN853 on day 1 of a 21-day cycle (once every 3 weeks dosing), with cycles repeated until patients experienced dose-limiting toxicity or progression. Dose escalation commenced in single-patient cohorts for the first 4 planned dose levels and then followed a standard 3 + 3 scheme. The primary objectives were to determine the maximum tolerated dose and the recommended phase 2 dose. Secondary objectives were to determine safety and tolerability, to characterize the pharmacokinetic profile, and to describe preliminary clinical activity. RESULTS: In total, 44 patients received treatment at doses escalating from 0.15 to 7.0 mg/kg. No meaningful drug accumulation was observed with the dosing regimen of once every 3 weeks. The most common treatment-related adverse events were fatigue, blurred vision, and diarrhea, the majority of which were grade 1 or 2. The dose-limiting toxicities observed were grade 3 hypophosphatemia (5.0 mg/kg) and grade 3 punctate keratitis (7.0 mg/kg). Two patients, both of whom were individuals with epithelial ovarian cancer, achieved confirmed tumor responses according to Response Evaluation Criteria in Solid Tumors 1.1, and each was a partial response. CONCLUSIONS: IMGN853 demonstrated a manageable safety profile and encouraging preliminary clinical activity, particularly in patients with ovarian cancer. The results establish a recommended phase 2 dosing of 6.0 mg/kg (based on adjusted ideal body weight) once every 3 weeks. Cancer 2017. © 2017 American Cancer Society. Cancer 2017;123:3080-7. © 2017 American Cancer Society.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Immunoconjugates/therapeutic use , Maytansine/analogs & derivatives , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Ovarian Epithelial , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Diarrhea/chemically induced , Disease Progression , Dose-Response Relationship, Drug , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Fatigue/chemically induced , Female , Humans , Hypophosphatemia/chemically induced , Keratitis/chemically induced , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Maximum Tolerated Dose , Maytansine/therapeutic use , Middle Aged , Neoplasms/pathology , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Vision Disorders/chemically induced
11.
Bioconjug Chem ; 28(5): 1371-1381, 2017 05 17.
Article in English | MEDLINE | ID: mdl-28388844

ABSTRACT

Antibody-drug conjugates (ADCs) are being actively pursued as a treatment option for cancer following the regulatory approval of brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla). ADCs consist of a cytotoxic agent conjugated to a targeting antibody through a linker. The two approved ADCs (and most ADCs now in the clinic that use a microtubule disrupting agent as the payload) are heterogeneous conjugates with an average drug-to-antibody ratio (DAR) of 3-4 (potentially ranging from 0 to 8 for individual species). Ado-trastuzumab emtansine employs DM1, a semisynthetic cytotoxic payload of the maytansinoid class, which is conjugated via lysine residues of the antibody to an average DAR of 3.5. To understand the effect of DAR on the preclinical properties of ADCs using maytansinoid cytotoxic agents, we prepared a series of conjugates with a cleavable linker (M9346A-sulfo-SPDB-DM4 targeting folate receptor α (FRα)) or an uncleavable linker (J2898A-SMCC-DM1 targeting the epidermal growth factor receptor (EGFR)) with varying DAR and evaluated their biochemical characteristics, in vivo stability, efficacy, and tolerability. For both formats, a series of ADCs with DARs ranging from low (average of ∼2 and range of 0-4) to very high (average of 10 and range of 7-14) were prepared in good yield with high monomer content and low levels of free cytotoxic agent. The in vitro potency consistently increased with increasing DAR at a constant antibody concentration. We then characterized the in vivo disposition of these ADCs. Pharmacokinetic analysis showed that conjugates with an average DAR below ∼6 had comparable clearance rates, but for those with an average DAR of ∼9-10, rapid clearance was observed. Biodistribution studies in mice showed that these 9-10 DAR ADCs rapidly accumulate in the liver, with maximum localization for this organ at 24-28% percentage injected dose per gram (%ID/g) compared with 7-10% for lower-DAR conjugates (all at 2-6 h post-injection). Our preclinical findings on tolerability and efficacy suggest that maytansinoid conjugates with DAR ranging from 2 to 6 have a better therapeutic index than conjugates with very high DAR (∼9-10). These very high DAR ADCs suffer from decreased efficacy, likely due to faster clearance. These results support the use of DAR 3-4 for maytansinoid ADCs but suggest that the exploration of lower or higher DAR may be warranted depending on the biology of the target antigen.


Subject(s)
Antibodies, Monoclonal/immunology , Antineoplastic Agents, Phytogenic/pharmacokinetics , Immunoconjugates/pharmacokinetics , Maytansine/pharmacokinetics , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Female , Humans , Immunoconjugates/pharmacology , KB Cells , Maytansine/pharmacology , Mice , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Neoplasia ; 18(12): 775-784, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27889646

ABSTRACT

Elevated folate receptor alpha (FRα) expression is characteristic of epithelial ovarian cancer (EOC), thus establishing this receptor as a candidate target for the development of novel therapeutics to treat this disease. Mirvetuximab soravtansine (IMGN853) is an antibody-drug conjugate (ADC) that targets FRα for tumor-directed delivery of the maytansinoid DM4, a potent agent that induces mitotic arrest by suppressing microtubule dynamics. Here, combinations of IMGN853 with approved therapeutics were evaluated in preclinical models of EOC. Combinations of IMGN853 with carboplatin or doxorubicin resulted in synergistic antiproliferative effects in the IGROV-1 ovarian cancer cell line in vitro. IMGN853 potentiated the cytotoxic activity of carboplatin via growth arrest and augmented DNA damage; cell cycle perturbations were also observed in cells treated with the IMGN853/doxorubicin combination. These benefits translated into improved antitumor activity in patient-derived xenograft models in vivo in both the platinum-sensitive (IMGN853/carboplatin) and platinum-resistant (IMGN853/pegylated liposomal doxorubicin) settings. IMGN853 co-treatment also improved the in vivo efficacy of bevacizumab in platinum-resistant EOC models, with combination regimens causing significant regressions and complete responses in the majority of tumor-bearing mice. Histological analysis of OV-90 ovarian xenograft tumors revealed that concurrent administration of IMGN853 and bevacizumab caused rapid disruption of tumor microvasculature and extensive necrosis, underscoring the superior bioactivity profile of the combination regimen. Overall, these demonstrations of combinatorial benefit conferred by the addition of the first FRα-targeting ADC to established therapies provide a compelling framework for the potential application of IMGN853 in the treatment of patients with advanced ovarian cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Folate Receptor 1/antagonists & inhibitors , Folate Receptor 1/metabolism , Immunoconjugates/pharmacology , Maytansine/analogs & derivatives , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Animals , Bevacizumab/pharmacology , Carboplatin/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Humans , Maytansine/pharmacology , Mice , Neovascularization, Pathologic/drug therapy , Ovarian Neoplasms/drug therapy , Platinum/pharmacology , Xenograft Model Antitumor Assays
13.
Bioconjug Chem ; 27(7): 1588-98, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27174129

ABSTRACT

Antibody-drug conjugates (ADCs) have become a widely investigated modality for cancer therapy, in part due to the clinical findings with ado-trastuzumab emtansine (Kadcyla). Ado-trastuzumab emtansine utilizes the Ab-SMCC-DM1 format, in which the thiol-functionalized maytansinoid cytotoxic agent, DM1, is linked to the antibody (Ab) via the maleimide moiety of the heterobifunctional SMCC linker. The pharmacokinetic (PK) data for ado-trastuzumab emtansine point to a faster clearance for the ADC than for total antibody. Cytotoxic agent release in plasma has been reported with nonmaytansinoid, cysteine-linked ADCs via thiol-maleimide exchange, for example, brentuximab vedotin. For Ab-SMCC-DM1 ADCs, however, the main catabolite reported is lysine-SMCC-DM1, the expected product of intracellular antibody proteolysis. To understand these observations better, we conducted a series of studies to examine the stability of the thiol-maleimide linkage, utilizing the EGFR-targeting conjugate, J2898A-SMCC-DM1, and comparing it with a control ADC made with a noncleavable linker that lacked a thiol-maleimide adduct (J2898A-(CH2)3-DM). We employed radiolabeled ADCs to directly measure both the antibody and the ADC components in plasma. The PK properties of the conjugated antibody moiety of the two conjugates, J2898A-SMCC-DM1 and J2898A-(CH2)3-DM (each with an average of 3.0 to 3.4 maytansinoid molecules per antibody), appear to be similar to that of the unconjugated antibody. Clearance values of the intact conjugates were slightly faster than those of the Ab components. Furthermore, J2898A-SMCC-DM1 clears slightly faster than J2898A-(CH2)3-DM, suggesting that there is a fraction of maytansinoid loss from the SMCC-DM1 ADC, possibly through a thiol-maleimide dependent mechanism. Experiments on ex vivo stability confirm that some loss of maytansinoid from Ab-SMCC-DM1 conjugates can occur via thiol elimination, but at a slower rate than the corresponding rate of loss reported for thiol-maleimide links formed at thiols derived by reduction of endogenous cysteine residues in antibodies, consistent with expected differences in thiol-maleimide stability related to thiol pKa. These findings inform the design strategy for future ADCs.


Subject(s)
Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Lysine/chemistry , Maleimides/chemistry , Maytansine/chemistry , Animals , Drug Stability , Mice , Structure-Activity Relationship
14.
Bioconjug Chem ; 26(11): 2261-78, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26355774

ABSTRACT

Antibody anilino maytansinoid conjugates (AaMCs) have been prepared in which a maytansinoid bearing an aniline group was linked through the aniline amine to a dipeptide, which in turn was covalently attached to a desired monoclonal antibody. Several such conjugates were prepared utilizing different dipeptides in the linkage including Gly-Gly, l-Val-l-Cit, and all four stereoisomers of the Ala-Ala dipeptide. The properties of AaMCs could be altered by the choice of dipeptide in the linker. Each of the AaMCs, except the AaMC bearing a d-Ala-d-Ala peptide linker, displayed more bystander killing in vitro than maytansinoid ADCs that utilize disulfide linkers. In mouse models, the anti-CanAg AaMC bearing a d-Ala-l-Ala dipeptide in the linker was shown to be more efficacious against heterogeneous HT-29 xenografts than maytansinoid ADCs that utilize disulfide linkers, while both types of the conjugates displayed similar tolerabilities.


Subject(s)
Aniline Compounds/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Immunoconjugates/chemistry , Maytansine/chemistry , Aniline Compounds/pharmacokinetics , Aniline Compounds/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Maytansine/pharmacokinetics , Maytansine/therapeutic use , Mice , Neoplasms/drug therapy
15.
Immunology ; 130(2): 231-42, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20201988

ABSTRACT

Adjuvants, including antibodies to tumour necrosis factor receptor superfamily members, augment immune responses. One member of this family, glucocorticoid-induced tumour necrosis factor receptor (GITR), is expressed at low levels on naive/resting T cells, B cells and macrophages, but at higher levels on T regulatory cells. The aim of this study was to determine the ability of a rat anti-mouse GITR monoclonal antibody, 2F8, to stimulate murine humoral and cellular immunity in a prime boost model with particular attention to posology and antigen-specific effects. 2F8 enhanced the humoral immune response to ovalbumin and haemagglutinin (HA) compared with controls and this enhancement was equal to or greater than that obtained in mice dosed with standard adjuvants. 2F8 F(ab')(2) fragments were as effective as intact antibody in boosting humoral immunity, indicating that FcR-mediated cross-linking of 2F8 is not required for efficacy. Moreover, the enhanced response was durable and antigen specific. Administration of 2F8 shifted the immune response towards a T helper type 1 response with significant enhancement of immunoglobulin G2a- and G2b-specific anti-HA antibodies, as well as enhanced cellular immunity as measured by ELISPOT. 2F8-treated mice also generated significantly more neutralizing antibodies to HA than control mice. Our findings show that anti-GITR is a robust, versatile adjuvant that, unlike commonly used adjuvants that primarily enhance humoral immunity, enhances both humoral and cellular immunity. These results support the continued development of anti-GITR for such indications as haematological and solid tumours, chronic viral infections, and as a vaccine adjuvant.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibodies, Monoclonal/pharmacology , B-Lymphocytes/immunology , Immunity, Humoral/drug effects , Macrophages/immunology , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , Th1 Cells/immunology , Animals , Antibodies, Monoclonal/immunology , Glucocorticoid-Induced TNFR-Related Protein , Hemagglutinins/immunology , Hemagglutinins/pharmacology , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/pharmacology , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Ovalbumin/pharmacology , Rats
16.
Cancer Biol Ther ; 8(7): 548-56, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19252416

ABSTRACT

Our previous studies showed that the depletion of the outer kinetochore protein hBub1 upon activation of spindle assembly checkpoint (SAC) primarily triggers early cell death mediated by p53 rather than aneuploidy. Here, we report that phosphorylation of p53 at Ser37 is critical for proapoptotic activity upon SAC activation. Furthermore, we show that p53 physically interacts with hBub1 at kinetochores in response to mitotic spindle damage suggesting a direct role for hBub1 in the suppression of p53 mediated cell death. This observation is further substantiated by the inhibition of p53 mediated transactivation of the proapoptotic target genes, PUMA and BAX, by hBub1 in SAC activated cells. In summary, our data from these and our previous studies strongly suggest that in response to SAC activation, hBub1 acts as a negative regulator of p53 mediated early cell death in a novel checkpoint pathway. On the translational medicine front, it is tempting to speculate that by disabling hBub1 in p53 proficient cancer cells, apoptosis may be induced as a therapeutic approach to eradicate the tumor cells.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Cycle/physiology , Cell Death/genetics , Cell Line, Tumor , Fluorescent Antibody Technique , HCT116 Cells , Humans , Immunoblotting , Immunoprecipitation , Nocodazole/pharmacology , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Spindle Apparatus/genetics , Tumor Suppressor Protein p53/genetics
17.
Cancer Biol Ther ; 8(7): 627-35, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19242126

ABSTRACT

It has been universally believed that spindle assembly checkpoint (SAC) proteins which include the kinetochore proteins are involved in monitoring the faithful segregation of sister chromatids during cell division and hence defects in these proteins result in aneuploidy. Furthermore, there are multiple sources of experimental data to suggest that a defect in p53 can also promote genomic instability leading to aneuploidy. Despite these observations, a molecular basis for the prevention of aneuploidy to maintain genomic integrity upon activation of SAC has largely remained elusive. In this report, we demonstrate a novel mechanism for the maintenance of a balance between cell survival and apoptosis upon activation of SAC. We found that depletion of the outer kinetochore protein hBub1 upon activation of SAC primarily triggers early cell death mediated by p53. This phenomenon is further supported by the upregulation of p53 downstream pro-apoptotic genes, BAX and PUMA as well as a corresponding increase in the cleavage products of PARP and caspase 3, markers of apoptosis, upon depletion of hBub1 in SAC activated cells. On the other hand, as expected, concomitant loss of both hBub1 and p53 resulted in disabling of the p53 mediated cell death pathway leading to the accumulation of cells with aneuploidy/polyploidy.


Subject(s)
Apoptosis/physiology , Protein Serine-Threonine Kinases/deficiency , Spindle Apparatus/metabolism , Tumor Suppressor Protein p53/metabolism , Aneuploidy , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Growth Processes/physiology , HCT116 Cells , Humans , Mad2 Proteins , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Spindle Apparatus/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Up-Regulation , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics
18.
Hum Mol Genet ; 15(21): 3132-45, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16984965

ABSTRACT

The variability in phenotypic presentations and the lack of consistency of genetic associations in mental illnesses remain a major challenge in molecular psychiatry. Recently, it has become increasingly clear that altered promoter DNA methylation could play a critical role in mediating differential regulation of genes and in facilitating short-term adaptation in response to the environment. Here, we report the investigation of the differential activity of membrane-bound catechol-O-methyltransferase (MB-COMT) due to altered promoter methylation and the nature of the contribution of COMT Val158Met polymorphism as risk factors for schizophrenia and bipolar disorder by analyzing 115 post-mortem brain samples from the frontal lobe. These studies are the first to reveal that the MB-COMT promoter DNA is frequently hypomethylated in schizophrenia and bipolar disorder patients, compared with the controls (methylation rate: 26 and 29 versus 60%; P=0.004 and 0.008, respectively), particularly in the left frontal lobes (methylation rate: 29 and 30 versus 81%; P=0.003 and 0.002, respectively). Quantitative gene-expression analyses showed a corresponding increase in transcript levels of MB-COMT in schizophrenia and bipolar disorder patients compared with the controls (P=0.02) with an accompanying inverse correlation between MB-COMT and DRD1 expression. Furthermore, there was a tendency for the enrichment of the Val allele of the COMT Val158Met polymorphism with MB-COMT hypomethylation in the patients. These findings suggest that MB-COMT over-expression due to promoter hypomethylation and/or hyperactive allele of COMT may increase dopamine degradation in the frontal lobe providing a molecular basis for the shared symptoms of schizophrenia and bipolar disorder.


Subject(s)
Bipolar Disorder/genetics , Catechol O-Methyltransferase/genetics , DNA Methylation , Frontal Lobe/metabolism , Promoter Regions, Genetic , Schizophrenia/genetics , Alcoholism/genetics , Amino Acid Substitution , Antipsychotic Agents/therapeutic use , Bipolar Disorder/drug therapy , Bipolar Disorder/metabolism , Case-Control Studies , CpG Islands/genetics , Epigenesis, Genetic , Genetic Predisposition to Disease , Heart Diseases/genetics , Humans , Polymorphism, Genetic , Receptors, Dopamine D1/genetics , Risk Factors , Schizophrenia/drug therapy , Schizophrenia/metabolism
19.
Clin Cancer Res ; 11(7): 2466-70, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15814621

ABSTRACT

Promoter DNA methylation status of six genes in samples derived from 27 bronchial epithelial cells and matching blood samples from 22 former/current smokers and five nonsmokers as well as 49 primary non-small cell lung cancer samples with corresponding blood controls was determined using methylation-specific PCR (MSP). Lung tumor tissues showed a significantly higher frequency of promoter DNA methylation in p16, MGMT, and DAPK (P < 0.05; Fisher's exact test). p16 promoter DNA methylation in tumors was observed at consistently higher levels when compared with all the other samples analyzed (P = 0.001; Fisher's exact test). ECAD and DAPK exhibited statistically insignificant differences in their levels of DNA methylation among the tumors and bronchial epithelial cells from the smokers. Interestingly, similar levels of methylation were observed in bronchial epithelial cells and corresponding blood from smokers for all four genes (ECAD, p16, MGMT, and DAPK) that showed smoking/lung cancer-associated methylation changes. In summary, our data suggest that targeted DNA methylation silencing of ECAD and DAPK occurs in the early stages and that of p16 and MGMT in the later stages of lung cancer progression. We also provide preliminary evidence that peripheral lymphocytes could potentially be used as a surrogate for bronchial epithelial cells to detect altered DNA methylation in smokers.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , DNA Methylation , Lung Neoplasms/genetics , Smoking/adverse effects , Apoptosis Regulatory Proteins , Bronchi/metabolism , Bronchi/pathology , Cadherins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Carcinoma, Non-Small-Cell Lung/etiology , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , DNA-Binding Proteins/genetics , Death-Associated Protein Kinases , Disease Progression , Gene Expression Regulation, Neoplastic , Glutathione S-Transferase pi , Glutathione Transferase/genetics , Humans , Isoenzymes/genetics , Lung Neoplasms/blood , Lung Neoplasms/etiology , Models, Biological , Neoplasm Staging , O(6)-Methylguanine-DNA Methyltransferase/genetics , Polymerase Chain Reaction , Precancerous Conditions/genetics , Promoter Regions, Genetic/genetics , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Smad8 Protein , Trans-Activators/genetics
20.
Cancer Res ; 65(5): 1664-9, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15753360

ABSTRACT

Dilution end point loss of heterozygosity (LOH) analysis, a novel approach for the analysis of LOH, was used to evaluate allelic losses with the use of 21 highly polymorphic microsatellite markers at nine chromosomal sites most frequently affected in smoking-related non-small cell lung cancers. Allelotyping was done for bronchial epithelial cells and matching blood samples from 23 former and current smokers and six nonsmokers as well as in 33 adenocarcinomas and 25 squamous cell carcinomas (SCC) and corresponding matching blood from smokers. Major conclusions from these studies are as follows: (a) LOH at chromosomal sites 8p, 9p, 11q, and 13q (P >0.05, Fisher's exact test) are targeted at the early stages, whereas LOH at 1p, 5q, 17p, and 18q (P <0.05, Fisher's exact test) occur at the later stages of non-small cell lung cancer progression; (b) LOH at 1p, 3p, 5q, 8p, 9p, 11q, 13q, 17p, and 18q occurs in over 45% of the tobacco smokers with SCC and adenocarcinoma; (c) compared with bronchial epithelial cells from smokers, there is a significantly higher degree of LOH at 1p, 5q, and 18q in adenocarcinoma and at 1p, 3p, and 17p in SCC (P <0.05, Fisher's exact test). We propose that lung cancer progression induced by tobacco smoke occurs in a series of target gene inactivations/activations in defined modules of a global network. The gatekeeper module consists of multiple alternate target genes, which is inclusive of but not limited to genes localized to chromosomal loci 8p, 9p, 11q, and 13q.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Chromosomes, Human/genetics , Loss of Heterozygosity , Lung Neoplasms/genetics , Smoking/adverse effects , Adenocarcinoma/blood , Adenocarcinoma/genetics , Aged , Alleles , Bronchi/metabolism , Bronchi/pathology , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/genetics , Case-Control Studies , DNA, Neoplasm/blood , DNA, Neoplasm/genetics , Dermatoglyphics , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Genetic Heterogeneity , Genotype , Humans , Lung Neoplasms/blood , Male , Microsatellite Repeats , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...