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 ; 22(2): 254-263, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36722141

ABSTRACT

Antibody-drug conjugates (ADC) delivering pyrrolobenzodiazepine (PBD) DNA cross-linkers are currently being evaluated in clinical trials, with encouraging results in Hodgkin and non-Hodgkin lymphomas. The first example of an ADC delivering a PBD DNA cross-linker (loncastuximab tesirine) has been recently approved by the FDA for the treatment of relapsed and refractory diffuse large B-cell lymphoma. There has also been considerable interest in mono-alkylating PBD analogs. We conducted a head-to-head comparison of a conventional PBD bis-imine and a novel PBD mono-imine. Key Mitsunobu chemistry allowed clean and convenient access to the mono-imine class. Extensive DNA-binding studies revealed that the mono-imine mediated a type of DNA interaction that is described as "pseudo cross-linking," as well as alkylation. The PBD mono-imine ADC demonstrated robust antitumor activity in mice bearing human tumor xenografts at doses 3-fold higher than those that were efficacious for the PBD bis-imine ADC. A single-dose toxicology study in rats demonstrated that the MTD of the PBD mono-alkylator ADC was approximately 3-fold higher than that of the ADC bearing a bis-imine payload, suggesting a comparable therapeutic index for this molecule. However, although both ADCs caused myelosuppression, renal toxicity was observed only for the bis-imine, indicating possible differences in toxicologic profiles that could influence tolerability and therapeutic index. These data show that mono-amine PBDs have physicochemical and pharmacotoxicologic properties distinct from their cross-linking analogs and support their potential utility as a novel class of ADC payload.


Subject(s)
Immunoconjugates , Lymphoma, Non-Hodgkin , Humans , Animals , Mice , Rats , Alkylation , DNA , Imines , Immunoconjugates/pharmacology
2.
Chem Commun (Camb) ; 59(13): 1841-1844, 2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36722863

ABSTRACT

Antibody-drug conjugates containing peroxide-cleavable arylboronic acid linkers are described, which target the high levels of reactive oxygen species (ROS) in cancer. The arylboronic acid linkers rapidly release a payload in the presence of hydrogen peroxide, but remain stable in plasma. Anti-HER2 and PD-L1 peroxide-cleavable ADCs exhibited potent cytotoxicity in vitro.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Immunoconjugates/pharmacology , Peroxides , Antineoplastic Agents/pharmacology , Hydrogen Peroxide , Acids
3.
Mol Cancer Ther ; 19(8): 1649-1659, 2020 08.
Article in English | MEDLINE | ID: mdl-32404408

ABSTRACT

Multiple myeloma is a hematologic cancer that disrupts normal bone marrow function and has multiple lines of therapeutic options, but is incurable as patients ultimately relapse. We developed a novel antibody-drug conjugate (ADC) targeting CS-1, a protein that is highly expressed on multiple myeloma tumor cells. The anti-CS-1 mAb specifically bound to cells expressing CS-1 and, when conjugated to a cytotoxic pyrrolobenzodiazepine payload, reduced the viability of multiple myeloma cell lines in vitro In mouse models of multiple myeloma, a single administration of the CS-1 ADC caused durable regressions in disseminated models and complete regression in a subcutaneous model. In an exploratory study in cynomolgus monkeys, the CS-1 ADC demonstrated a half-life of 3 to 6 days; however, no highest nonseverely toxic dose was achieved, as bone marrow toxicity was dose limiting. Bone marrow from dosed monkeys showed reductions in progenitor cells as compared with normal marrow. In vitro cell killing assays demonstrated that the CS-1 ADC substantially reduced the number of progenitor cells in healthy bone marrow, leading us to identify previously unreported CS-1 expression on a small population of progenitor cells in the myeloid-erythroid lineage. This finding suggests that bone marrow toxicity is the result of both on-target and off-target killing by the ADC.


Subject(s)
Antibodies, Monoclonal/chemistry , Antineoplastic Agents/pharmacology , Benzodiazepines/chemistry , Immunoconjugates/pharmacology , Membrane Proteins/antagonists & inhibitors , Microfilament Proteins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Pyrroles/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis , Cell Proliferation , Drug Evaluation, Preclinical , Female , Humans , Immunoconjugates/chemistry , Macaca fascicularis , Membrane Proteins/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Microfilament Proteins/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
PLoS Negl Trop Dis ; 13(5): e0007373, 2019 05.
Article in English | MEDLINE | ID: mdl-31120889

ABSTRACT

Infections of humans and livestock with African trypanosomes are treated with drugs introduced decades ago that are not always fully effective and often have severe side effects. Here, the trypanosome haptoglobin-haemoglobin receptor (HpHbR) has been exploited as a route of uptake for an antibody-drug conjugate (ADC) that is completely effective against Trypanosoma brucei in the standard mouse model of infection. Recombinant human anti-HpHbR monoclonal antibodies were isolated and shown to be internalised in a receptor-dependent manner. Antibodies were conjugated to a pyrrolobenzodiazepine (PBD) toxin and killed T. brucei in vitro at picomolar concentrations. A single therapeutic dose (0.25 mg/kg) of a HpHbR antibody-PBD conjugate completely cured a T. brucei mouse infection within 2 days with no re-emergence of infection over a subsequent time course of 77 days. These experiments provide a demonstration of how ADCs can be exploited to treat protozoal diseases that desperately require new therapeutics.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antiprotozoal Agents/administration & dosage , Benzodiazepines/administration & dosage , Pyrroles/administration & dosage , Trypanosomiasis, African/drug therapy , Animals , Antibodies, Monoclonal/chemistry , Antiprotozoal Agents/chemistry , Benzodiazepines/chemistry , Female , Humans , Mice , Mice, Inbred BALB C , Pyrroles/chemistry , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/parasitology
5.
Bioconjug Chem ; 30(4): 1232-1243, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30912649

ABSTRACT

Despite some clinical success with antibody-drug conjugates (ADCs) in patients with solid tumors and hematological malignancies, improvements in ADC design are still desirable due to the narrow therapeutic window of these compounds. Tumor-targeting antibody fragments have distinct advantages over monoclonal antibodies, including more rapid tumor accumulation and enhanced penetration, but are subject to rapid clearance. Half-life extension technologies such as PEGylation and albumin-binding domains (ABDs) have been widely used to improve the pharmacokinetics of many different types of biologics. PEGylation improves pharmacokinetics by increasing hydrodynamic size to reduce renal clearance, whereas ABDs extend half-life via FcRn-mediated recycling. In this study, we used an anti-oncofetal antigen 5T4 diabody conjugated with a highly potent cytotoxic pyrrolobenzodiazepine (PBD) warhead to assess and compare the effects of PEGylation and albumin binding on the in vivo efficacy of antibody fragment drug conjugates. Conjugation of 2× PEG20K to a diabody improved half-life from 40 min to 33 h, and an ABD-diabody fusion protein exhibited a half-life of 45 h in mice. In a xenograft model of breast cancer MDA-MB-436, the ABD-diabody-PBD showed greater tumor growth suppression and better tolerability than either PEG-diabody-PBD or diabody-PBD. These results suggest that the mechanism of half-life extension is an important consideration for designing cytotoxic antitumor agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunoconjugates/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Binding, Competitive , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enzyme-Linked Immunosorbent Assay , Female , Half-Life , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Mice , Mice, Nude , Polyethylene Glycols/chemistry , Xenograft Model Antitumor Assays
6.
J Org Chem ; 84(8): 4830-4836, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30602115

ABSTRACT

An analysis of Antibody-Drug Conjugate Payload manufacturing has revealed that the majority of the cost is associated with the use of high-containment facilities for the latter stages of the synthesis. To make a significant reduction in the Cost of Goods (CoGs), a new approach to route design has been introduced which focuses on minimizing the number of steps that require high containment. This approach has been exemplified in a new synthesis of tesirine, including the first application of a ring-closing copper(I)/TEMPO aerobic oxidation to the pyrrolobenzodiazepine ring system, affording a 60% reduction in CoGs.


Subject(s)
Benzodiazepines/chemical synthesis , Drug Design , Immunoconjugates/chemistry , Pyrroles/chemical synthesis , Benzodiazepines/chemistry , Cyclization , Molecular Structure , Pyrroles/chemistry
7.
Clin Cancer Res ; 24(24): 6570-6582, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30131388

ABSTRACT

PURPOSE: Antibody-drug conjugates (ADC) utilizing noncleavable linker drugs have been approved for clinical use, and several are in development targeting solid and hematologic malignancies including multiple myeloma. Currently, there are no reliable biomarkers of activity for these ADCs other than presence of the targeted antigen. We observed that certain cell lines are innately resistant to such ADCs, and sought to uncover the underlying mechanism of resistance. EXPERIMENTAL DESIGN: The expression of 43 lysosomal membrane target genes was evaluated in cell lines resistant to ADCs bearing the noncleavable linker, pyrrolobenzodiazepine payload SG3376, in vitro. The functional relevance of SLC46A3, a lysosomal transporter of noncleavable ADC catabolites whose expression uniquely correlated with SG3376 resistance, was assessed using EPHA2-, HER2-, and BCMA-targeted ADCs and isogenic cells overexpressing or genetically inactivated for SLC46A3. SLC46A3 expression was also examined in patient-derived xenograft and in vitro models of acquired T-DM1 resistance and multiple myeloma bone marrow samples by RT-PCR. RESULTS: Loss of SLC46A3 expression was found to be a mechanism of innate and acquired resistance to ADCs bearing DM1 and SG3376. Sensitivity was restored in refractory lines upon introduction of SLC46A3, suggesting that expression of SLC46A3 may be more predictive of activity than target antigen levels alone. Interrogation of primary multiple myeloma samples indicated a range of SLC46A3 expression, including samples with undetectable levels like multiple myeloma cell lines resistant to BCMA-targeting DM1 and SG3376 ADCs. CONCLUSIONS: Our findings support SLC46A3 as a potential patient selection biomarker with immediate relevance to clinical trials involving these ADCs.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Benzodiazepines/pharmacology , Biomarkers , Immunoconjugates/pharmacology , Maytansine/pharmacology , Pyrroles/pharmacology , Animals , Antineoplastic Agents, Immunological/chemistry , Benzodiazepines/chemistry , Cell Line, Tumor , Disease Models, Animal , Gene Expression , Gene Silencing , Humans , Immunoconjugates/chemistry , Maytansine/chemistry , Melanoma, Experimental , Mice , Pyrroles/chemistry , Xenograft Model Antitumor Assays
8.
Sci Rep ; 8(1): 10479, 2018 Jul 11.
Article in English | MEDLINE | ID: mdl-29992976

ABSTRACT

Synthetic pyrrolobenzodiazepine (PBD) dimers, where two PBD monomers are linked through their aromatic A-ring phenolic C8-positions via a flexible propyldioxy tether, are highly efficient DNA minor groove cross-linking agents with potent cytotoxicity. PBD dimer SG3199 is the released warhead component of the antibody-drug conjugate (ADC) payload tesirine (SG3249), currently being evaluated in several ADC clinical trials. SG3199 was potently cytotoxic against a panel of human solid tumour and haematological cancer cell lines with a mean GI50 of 151.5 pM. Cells defective in DNA repair protein ERCC1 or homologous recombination repair showed increased sensitivity to SG3199 and the drug was only moderately susceptible to multidrug resistance mechanisms. SG3199 was highly efficient at producing DNA interstrand cross-links in naked linear plasmid DNA and dose-dependent cross-linking was observed in cells. Cross-links formed rapidly in cells and persisted over 36 hours. Following intravenous (iv) administration to rats SG3199 showed a very rapid clearance with a half life as short as 8 minutes. These combined properties of cytotoxic potency, rapid formation and persistence of DNA interstrand cross-links and very short half-life contribute to the emerging success of SG3199 as a warhead in clinical stage ADCs.


Subject(s)
Antineoplastic Agents/chemistry , Benzodiazepines/pharmacokinetics , Immunotoxins/chemistry , Pyrroles/pharmacokinetics , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Benzodiazepines/therapeutic use , Cell Line, Tumor , Cross-Linking Reagents , DNA/metabolism , DNA Repair , Dimerization , Drug Screening Assays, Antitumor , Humans , Pyrroles/therapeutic use , Rats
9.
Blood ; 131(10): 1094-1105, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29298756

ABSTRACT

Human CD19 antigen is a 95-kDa type I membrane glycoprotein in the immunoglobulin superfamily whose expression is limited to the various stages of B-cell development and differentiation and is maintained in the majority of B-cell malignancies, including leukemias and non-Hodgkin lymphomas of B-cell origin. Coupled with its differential and favorable expression profile, CD19 has rapid internalization kinetics and is not shed into the circulation, making it an ideal target for the development of antibody-drug conjugates (ADCs) to treat B-cell malignancies. ADCT-402 (loncastuximab tesirine) is a novel CD19-targeted ADC delivering SG3199, a highly cytotoxic DNA minor groove interstrand crosslinking pyrrolobenzodiazepine (PDB) dimer warhead. It showed potent and highly targeted in vitro cytotoxicity in CD19-expressing human cell lines. ADCT-402 was specifically bound, internalized, and trafficked to lysosomes in CD19-expressing cells and, following release of the PBD warhead, resulted in formation of DNA crosslinks that persisted for 36 hours. Bystander killing of CD19- cells by ADCT-402 was also observed. In vivo, single doses of ADCT-402 resulted in highly potent, dose-dependent antitumor activity in several subcutaneous and disseminated human tumor models with marked superiority to comparator ADCs delivering tubulin inhibitors. Dose-dependent DNA crosslinks and γ-H2AX DNA damage response were measured in tumors by 24 hours after single dose administration, whereas matched peripheral blood mononuclear cells showed no evidence of DNA damage. Pharmacokinetic analysis in rat and cynomolgus monkey showed excellent stability and tolerability of ADCT-402 in vivo. Together, these impressive data were used to support the clinical testing of this novel ADC in patients with CD19-expressing B-cell malignancies.


Subject(s)
Antigens, CD19/biosynthesis , Antineoplastic Agents , Gene Expression Regulation, Leukemic , Immunoconjugates , Leukemia, B-Cell , Lymphoma, Non-Hodgkin , Neoplasm Proteins/biosynthesis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/metabolism , Leukemia, B-Cell/pathology , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/pathology , Lysosomes/metabolism , Lysosomes/pathology
10.
Clin Cancer Res ; 23(19): 5858-5868, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28630216

ABSTRACT

Purpose: To use preclinical models to identify a dosing schedule that improves tolerability of highly potent pyrrolobenzodiazepine dimers (PBDs) antibody drug conjugates (ADCs) without compromising antitumor activity.Experimental Design: A series of dose-fractionation studies were conducted to investigate the pharmacokinetic drivers of safety and efficacy of PBD ADCs in animal models. The exposure-activity relationship was investigated in mouse xenograft models of human prostate cancer, breast cancer, and gastric cancer by comparing antitumor activity after single and fractionated dosing with tumor-targeting ADCs conjugated to SG3249, a potent PBD dimer. The exposure-tolerability relationship was similarly investigated in rat and monkey toxicology studies by comparing tolerability, as assessed by survival, body weight, and organ-specific toxicities, after single and fractionated dosing with ADCs conjugated to SG3249 (rats) or SG3400, a structurally related PBD (monkeys).Results: Observations of similar antitumor activity in mice treated with single or fractionated dosing suggests that antitumor activity of PBD ADCs is more closely related to total exposure (AUC) than peak drug concentrations (Cmax). In contrast, improved survival and reduced toxicity in rats and monkeys treated with a fractionated dosing schedule suggests that tolerability of PBD ADCs is more closely associated with Cmax than AUC.Conclusions: We provide the first evidence that fractionated dosing can improve preclinical tolerability of at least some PBD ADCs without compromising efficacy. These findings suggest that preclinical exploration of dosing schedule could be an important clinical strategy to improve the therapeutic window of highly potent ADCs and should be investigated further. Clin Cancer Res; 23(19); 5858-68. ©2017 AACR.


Subject(s)
Benzodiazepines/administration & dosage , Breast Neoplasms/drug therapy , Immunoconjugates/administration & dosage , Prostatic Neoplasms/drug therapy , Pyrroles/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Benzodiazepines/chemistry , Benzodiazepines/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Haplorhini , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Male , Mice , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Pyrroles/chemistry , Pyrroles/immunology , Rats , Therapeutic Index , Trastuzumab/administration & dosage , Trastuzumab/immunology , Xenograft Model Antitumor Assays
11.
Bioorg Med Chem Lett ; 27(5): 1154-1158, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28188066

ABSTRACT

A novel pyrrolobenzodiazepine dimer payload, SG3227, was rationally designed based on the naturally occurring antitumour compound sibiromycin. SG3227 was synthesized from a dimeric core in an efficient fashion. An unexpected room temperature Diels-Alder reaction occurred during the final step of the synthesis and was circumvented by use of an iodoacetamide conjugation moiety in place of a maleimide. The payload was successfully conjugated to trastuzumab and the resulting ADC exhibited potent activity against a HER2-expressing human cancer cell line in vitro.


Subject(s)
Aminoglycosides/chemistry , Antineoplastic Agents/chemistry , Benzodiazepines/chemistry , Immunoconjugates/chemistry , Cell Line, Tumor , Drug Evaluation, Preclinical , Humans , In Vitro Techniques
12.
Antibodies (Basel) ; 6(4)2017 Nov 28.
Article in English | MEDLINE | ID: mdl-31548535

ABSTRACT

Thiosuccinimide-linked antibody-drug conjugates (ADCs) are susceptible to drug loss over time due to a retro-Michael reaction, which can be prevented by selecting stable conjugation positions or hydrolysis of the thiosuccinimide. Here, we investigate pyrrolobenzodiazepine (PBD) ADC drug-linkers equipped with N-phenyl maleimide functionality for stable thiol conjugation via thiosuccinimide hydrolysis. Two PBD drug-linker formats (enzyme-cleavable and non-cleavable) were evaluated following site-specific conjugation to an engineered cysteine incorporated at position T289, which is known to be unstable for N-alkyl maleimide conjugates. N-phenyl maleimide PBDs conjugated to antibodies with similar efficiencies as N-alkyl maleimide PBDs and enhanced thiosuccinimide hydrolysis for N-phenyl maleimide PBDs was confirmed by mass spectrometry, capillary isoelectric focusing, and a SYPRO Orange dye binding assay. All of the PBD ADCs were highly potent in vitro regardless of maleimide- or linker-type, exhibiting low pM EC50 values. Thiol conjugation to N-phenyl maleimide PBD minimized the retro-Michael reaction in both rat and mouse serum. However, cleavage of the valine-alanine dipeptide in mouse serum for ADCs containing cleavable drug-linker led to drug loss regardless of maleimide type, which impacted ADC potency in tumor growth inhibition studies that were conducted in mouse models. Therapeutic improvement in mouse tumor models was realized for ADCs prepared with non-cleavable PBD drug-linkers that were conjugated through N-phenyl maleimide, where a stronger tumor growth inhibition (TGI) response was achieved when compared to the analogous N-alkyl maleimide drug-linker ADC. Altogether, our findings highlight the stability and efficacy benefits of N-phenyl maleimide functionality for ADCs that are produced with thiol-maleimide conjugation chemistry.

13.
ACS Med Chem Lett ; 7(11): 983-987, 2016 Nov 10.
Article in English | MEDLINE | ID: mdl-27882195

ABSTRACT

Pyrrolobenzodiazepine dimers are an emerging class of warhead in the field of antibody-drug conjugates (ADCs). Tesirine (SG3249) was designed to combine potent antitumor activity with desirable physicochemical properties such as favorable hydrophobicity and improved conjugation characteristics. One of the reactive imines was capped with a cathepsin B-cleavable valine-alanine linker. A robust synthetic route was developed to allow the production of tesirine on clinical scale, employing a flexible, convergent strategy. Tesirine was evaluated in vitro both in stochastic and engineered ADC constructs and was confirmed as a potent and versatile payload. The conjugation of tesirine to anti-DLL3 rovalpituzumab has resulted in rovalpituzumab-tesirine (Rova-T), currently under evaluation for the treatment of small cell lung cancer.

14.
Mol Cancer Ther ; 15(11): 2709-2721, 2016 11.
Article in English | MEDLINE | ID: mdl-27535974

ABSTRACT

Despite the many advances in the treatment of hematologic malignancies over the past decade, outcomes in refractory lymphomas remain poor. One potential strategy in this patient population is the specific targeting of IL2R-α (CD25), which is overexpressed on many lymphoma and leukemic cells, using antibody-drug conjugates (ADC). ADCT-301 is an ADC composed of human IgG1 HuMax-TAC against CD25, stochastically conjugated through a dipeptide cleavable linker to a pyrrolobenzodiazepine (PBD) dimer warhead with a drug-antibody ratio (DAR) of 2.3. ADCT-301 binds human CD25 with picomolar affinity. ADCT-301 has highly potent and selective cytotoxicity against a panel of CD25-expressing human lymphoma cell lines. Once internalized, the released warhead binds in the DNA minor groove and exerts its potent cytotoxic action via the formation of DNA interstrand cross-links. A strong correlation between loss of viability and DNA cross-link formation is demonstrated. DNA damage persists, resulting in phosphorylation of histone H2AX, cell-cycle arrest in G2-M, and apoptosis. Bystander killing of CD25-negative cells by ADCT-301 is also observed. In vivo, a single dose of ADCT-301 results in dose-dependent and targeted antitumor activity against both subcutaneous and disseminated CD25-positive lymphoma models. In xenografts of Karpas 299, which expressed both CD25 and CD30, marked superiority over brentuximab vedotin (Adcetris) is observed. Dose-dependent increases in DNA cross-linking, γ-H2AX, and PBD payload staining were observed in tumors in vivo indicating a role as relevant pharmacodynamic assays. Together, these data support the clinical testing of this novel ADC in patients with CD25-expressing tumors. Mol Cancer Ther; 15(11); 2709-21. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Benzodiazepines , Hematologic Neoplasms/metabolism , Immunoconjugates/pharmacology , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Pyrroles , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Apoptosis/genetics , Benzodiazepines/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Histones/metabolism , Humans , Immunoconjugates/chemistry , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Pyrroles/chemistry , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Bioconjug Chem ; 26(11): 2249-60, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26332743

ABSTRACT

Antibody-drug conjugates (ADC) have emerged as potent antitumor drugs that provide increased efficacy, specificity, and tolerability over chemotherapy for the treatment of cancer. ADCs generated by targeting cysteines and lysines on the antibody have shown efficacy, but these products are heterogeneous, and instability may limit their dosing. Here, a novel technology is described that enables site-specific conjugation of toxins to antibodies using chemistry to produce homogeneous, potent, and highly stable conjugates. We have developed a cell-based mammalian expression system capable of site-specific integration of a non-natural amino acid containing an azide moiety. The azide group enables click cycloaddition chemistry that generates a stable heterocyclic triazole linkage. Antibodies to Her2/neu were expressed to contain N6-((2-azidoethoxy)carbonyl)-l-lysine at four different positions. Each site allowed over 95% conjugation efficacy with the toxins auristatin F or a pyrrolobenzodiazepine (PBD) dimer to generate ADCs with a drug to antibody ratio of >1.9. The ADCs were potent and specific in in vitro cytotoxicity assays. An anti Her2/neu conjugate demonstrated stability in vivo and a PBD containing ADC showed potent efficacy in a mouse tumor xenograph model. This technology was extended to generate fully functional ADCs with four toxins per antibody. The high stability of the azide-alkyne linkage, combined with the site-specific nature of the expression system, provides a means for the generation of ADCs with optimized pharmacokinetic, biological, and biophysical properties.


Subject(s)
Amino Acids/chemistry , Azides/chemistry , Click Chemistry , Cycloaddition Reaction , Immunoconjugates/chemistry , Amino Acids/genetics , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Azides/metabolism , Humans , Immunoconjugates/genetics , Immunoconjugates/immunology , Immunoconjugates/therapeutic use , Male , Mice , Neoplasms/drug therapy , Protein Engineering , Rats, Sprague-Dawley , Receptor, ErbB-2/immunology
16.
Bioconjug Chem ; 24(7): 1256-63, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23808985

ABSTRACT

A highly cytotoxic DNA cross-linking pyrrolobenzodiazepine (PBD) dimer with a valine-alanine dipeptide linker was conjugated to the anti-CD70 h1F6 mAb either through endogenous interchain cysteines or, site-specifically, through engineered cysteines at position 239 of the heavy chains. The h1F6239C-PBD conjugation strategy proved to be superior to interchain cysteine conjugation, affording an antibody-drug conjugate (ADC) with high uniformity in drug-loading and low levels of aggregation. In vitro cytotoxicity experiments demonstrated that the h1F6239C-PBD was potent and immunologically specific on CD70-positive renal cell carcinoma (RCC) and non-Hodgkin lymphoma (NHL) cell lines. The conjugate was resistant to drug loss in plasma and in circulation, and had a pharmacokinetic profile closely matching that of the parental h1F6239C antibody capped with N-ethylmaleimide (NEM). Evaluation in CD70-positive RCC and NHL mouse xenograft models showed pronounced antitumor activities at single or weekly doses as low as 0.1 mg/kg of ADC. The ADC was tolerated at 2.5 mg/kg. These results demonstrate that PBDs can be effectively used for antibody-targeted therapy.


Subject(s)
Benzodiazepines/chemistry , CD27 Ligand/chemistry , Immunoconjugates/pharmacology , Animals , Dimerization , Drug Design , Female , Half-Life , Immunoconjugates/chemistry , Mice , Mice, Inbred BALB C
17.
Cancer Res ; 70(17): 6849-58, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20660714

ABSTRACT

The pyrrolobenzodiazepines (PBD) are naturally occurring antitumor antibiotics, and a PBD dimer (SJG-136, SG2000) is in phase II trials. Many potent PBDs contain a C2-endo-exo unsaturated motif associated with the pyrrolo C-ring. The novel compound SG2202 is a PBD dimer containing this motif. SG2285 is a water-soluble prodrug of SG2202 in which two bisulfite groups inactivate the PBD N10-C11 imines. Once the bisulfites are eliminated, the imine moieties can bind covalently in the DNA minor groove, forming an interstrand cross-link. The mean in vitro cytotoxic potency of SG2285 against human tumor cell lines is GI(50) 20 pmol/L. SG2285 is highly efficient at producing DNA interstrand cross-links in cells, but they form more slowly than those produced by SG2202. Cellular sensitivity to SG2285 was primarily dependent on ERCC1 and homologous recombination repair. In primary B-cell chronic lymphocytic leukemia samples, the mean LD(50) was significantly lower than in normal age-matched B and T lymphocytes. Antitumor activity was shown in several human tumor xenograft models, including ovarian, non-small cell lung, prostate, pancreatic, and melanoma, with cures obtained in the latter model with a single dose. Further, in an advanced-stage colon model, SG2285 administered either as a single dose, or in two repeat dose schedules, was superior to irinotecan. Our findings define SG2285 as a highly active cytotoxic compound with antitumor properties desirable for further development.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Prodrugs/pharmacology , Animals , Benzodiazepinones/pharmacology , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Female , Humans , Mice , Neoplasms/drug therapy , Pyrroles/pharmacology , Xenograft Model Antitumor Assays
18.
J Med Chem ; 53(7): 2927-41, 2010 Apr 08.
Article in English | MEDLINE | ID: mdl-20218628

ABSTRACT

A comprehensive SAR investigation of the C2-position of pyrrolo[2,1-c][1,4]benzodiazepine (PBD) monomer antitumor agents is reported, establishing the molecular requirements for optimal in vitro cytotoxicity and DNA-binding affinity. Both carbocyclic and heterocyclic C2-aryl substituents have been studied ranging from single aryl rings to fused ring systems, and also styryl substituents, establishing across a library of 80 analogues that C2-aryl and styryl substituents significantly enhance both DNA-binding affinity and in vitro cytotoxicity, with a correlation between the two. The optimal C2-grouping for both DNA-binding affinity and cytotoxicity was found to be the C2-quinolinyl moiety which, according to molecular modeling, is due to the overall fit of the molecule in the DNA minor groove, and potential specific contacts with functional groups in the floor and walls of the groove. This analogue (14l) was shown to delay tumor growth in a HCT-116 (bowel) human tumor xenograft model.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Base Sequence , Benzodiazepines/chemical synthesis , Benzodiazepines/metabolism , Cattle , Cell Line, Tumor , DNA/chemistry , DNA/genetics , DNA/metabolism , Female , Humans , Imines/chemistry , Mice , Models, Molecular , Nucleic Acid Conformation , Nucleic Acid Denaturation/drug effects , Pyrroles/chemical synthesis , Pyrroles/metabolism , Stereoisomerism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
19.
Bioorg Med Chem Lett ; 19(22): 6463-6, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19811912

ABSTRACT

A prodrug form (17) of a novel C2/C2'-aryl-substituted pyrrolobenzodiazepine (PBD) dimer (16) has been synthesized by introducing sodium bisulfite groups to the C11/C11'-positions of the parent bis-imine. The prodrug form is highly water soluble, stable in aqueous conditions, and the rate of DNA cross-link formation is much slower compared to the parent bis-imine.


Subject(s)
Benzodiazepinones/chemical synthesis , DNA/drug effects , Drug Resistance, Neoplasm/drug effects , Prodrugs , Pyrroles/chemical synthesis , Animals , Benzodiazepines/pharmacology , Benzodiazepinones/chemistry , Benzodiazepinones/pharmacology , Cell Line, Tumor , Computer Simulation , DNA/metabolism , Drug Design , Drug Screening Assays, Antitumor , Epoxy Compounds/pharmacology , Humans , Models, Molecular , Pyrroles/chemistry , Pyrroles/pharmacology , Solubility/drug effects , Stereoisomerism , Structure-Activity Relationship
20.
Bioorg Med Chem Lett ; 18(6): 2073-7, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18272367

ABSTRACT

A novel sequence-selective extended PBD dimer 4 has been synthesized that binds with high affinity to an interstrand cross-linking site spanning 11 DNA base pairs. Despite its molecular weight (984.07) and length, the molecule has significant DNA interstrand cross-linking potency (approximately 100-fold greater than the clinically used agent melphalan) and sub-micromolar cytotoxicity in a number of tumour cell lines, suggesting that it readily penetrates cellular and nuclear membranes to reach its DNA target.


Subject(s)
Benzodiazepines/chemical synthesis , Benzodiazepines/pharmacology , Cross-Linking Reagents/chemical synthesis , Cross-Linking Reagents/pharmacology , DNA/metabolism , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Base Sequence , Benzodiazepines/chemistry , Cross-Linking Reagents/chemistry , DNA Footprinting , Dimerization , Humans , K562 Cells , Molecular Sequence Data , Molecular Structure , Pyrroles/chemistry , Sequence Homology, Nucleic Acid , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL