Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 93
1.
Toxicol Appl Pharmacol ; 485: 116886, 2024 Apr.
Article En | MEDLINE | ID: mdl-38452946

Despite extensive preclinical testing, cancer therapeutics can result in unanticipated toxicity to non-tumor tissue in patients. These toxicities may pass undetected in preclinical experiments due to modeling limitations involving poor biomimicry of 2-dimensional in vitro cell cultures and due to lack of interspecies translatability in in vivo studies. Instead, primary cells can be grown into miniature 3-dimensional structures that recapitulate morphological and functional aspects of native tissue, termed "organoids." Here, human bronchioalveolar organoids grown from primary alveolar epithelial cells were employed to model lung epithelium and investigate off-target toxicities associated with antibody-drug conjugates (ADCs). ADCs with three different linker-payload combinations (mafodotin, vedotin, and deruxtecan) were tested in bronchioalveolar organoids generated from human, rat, and nonhuman primate lung cells. Organoids demonstrated antibody uptake and changes in viability in response to ADC exposure that model in vivo drug sensitivity. RNA sequencing identified inflammatory activation in bronchioalveolar cells in response to deruxtecan. Future studies will explore specific cell populations involved in interstitial lung disease and incorporate immune cells to the culture.


Immunoconjugates , Organoids , Organoids/drug effects , Organoids/pathology , Animals , Immunoconjugates/toxicity , Humans , Rats , Drug Evaluation, Preclinical/methods , Macaca fascicularis , Cells, Cultured , Toxicity Tests/methods , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/pathology
2.
Drug Chem Toxicol ; 46(1): 24-34, 2023 Jan.
Article En | MEDLINE | ID: mdl-34839761

Mesothelin (MSLN) is an attractive therapeutic target for antibody drug conjugates (ADC) because of significant differences in expression pattern between diseased and normal tissues. RC88-ADC is a novel ADC, targeting MSLN, and inhibits tumor growth significantly in mice xenograft models. We performed an 11-week repeated dose toxicity study of RC88-ADC via intravenous injection in Cynomolgus Monkeys with an 8-Week recovery period according to International Conference on Harmonization (ICH) S9 and S6(R1). RC88-ADC was administered to groups of 5 male and 5 female monkeys at dose levels of 2.5, 5, and 10 mg/kg/2 weeks, meanwhile vehicle, naked antibody, small molecule groups were set up as the control. 4 animals died in 10 mg/kg group of RC88-ADC. The clinical symptoms mainly included ocular toxicity, weight loss and food intake decrease in the middle and high dose groups of RC88-ADC. RC88-ADC caused dose-related reversible myelosuppression, manifested as hematologic toxicity, which was consistent with the small molecule toxicity profile of its coupling. The highest non-severely toxic dose of RC88-ADC was 5 mg/kg in monkeys after repeated dosing. Nonetheless, the integrated analysis showed that RC88-ADC demonstrated an acceptable safety profile and provided an improved treatment window. These results pave the way for further investigation of RC88-ADC in humans.


Immunoconjugates , Neoplasms , Humans , Mice , Male , Female , Animals , Immunoconjugates/toxicity , Mesothelin , Neoplasms/drug therapy , Oligopeptides/therapeutic use , Cell Line, Tumor , Xenograft Model Antitumor Assays
3.
Analyst ; 147(23): 5386-5394, 2022 Nov 21.
Article En | MEDLINE | ID: mdl-36285914

Ligand-targeted drugs (LTDs) such as antibody-drug conjugates (ADCs) are currently attracting great attention as an alternative class of therapeutics to conventional chemotherapy for the clinical treatment of cancer. The linker is one of important factors determining the efficacy and toxicity of LTDs. The linker for LTDs should have enough stability during blood circulation, effectively release the payload, and leave no polar moieties in the released payload. However, the drug release activity and plasma stability of cleavable linkers are generally evaluated by complex and sophisticated in vivo techniques containing LC-MS, and the designing of new clinically applicable linkers remains a challenge. In this work, leucine aminopeptidase (LAP)-responsive fluorescent probes were designed as a simple preliminary model to verify whether a peptidase-responsive fluorescent probe can be used as a facile tool for the development of cleavable linkers although LAP is an exopeptidase and can't be a real target for cleavable linkers. LAP-responsive fluorescent probes were prepared by conjugation of a leucine to several xanthene fluorophores through a few linkages with a p-aminobenzyl spacer. The stability tests, kinetic study and live cell imaging of LAP-responsive activatable fluorescent probes demonstrated that the chemical stability and intrinsic activity of the linker for the release of drug can be easily evaluated by a fluorogenic assay. The ex vivo plasma stability test using mice suggested that an enzyme-responsive activatable fluorescent probe can be used as a feasible platform to evaluate the plasma stability of cleavable linkers during blood circulation.


Fluorescent Dyes , Immunoconjugates , Mice , Animals , Fluorescent Dyes/toxicity , Leucyl Aminopeptidase , Immunoconjugates/toxicity , Xanthenes , Drug Delivery Systems
4.
J Ocul Pharmacol Ther ; 38(7): 471-480, 2022 09.
Article En | MEDLINE | ID: mdl-35537481

Purpose: To characterize the ocular toxicity of an antibody-drug conjugate (ADC), depatuxizumab mafodotin (Depatux-m), in nonclinical species and to evaluate the effects of drug-antibody ratios (DARs), variations of the ADC construct, and potential methods for mitigation of the corneal toxicity. Depatux-m contains the potent cytotoxic agent monomethyl auristatin F as the ADC payload. Methods: Depatux-m was administered intravenously to cynomolgus monkeys at doses up to 30 mg/kg and to mice up to 100 mg/kg. Ocular toxicity was evaluated by clinical ophthalmic examinations and histopathology. Potential mitigation was tested through agents to block target engagement and multiple topical ophthalmic treatments (antioxidant, vasoconstrictor, tear stimulant). Results: Effects primarily involved corneal epithelium and were dose-dependent with respect to onset, severity, and time to reversal in both monkeys and mice. On slit lamp biomicroscopy, the initial effect in monkeys was superficial multifocal punctate opacities (granularity), which migrated axially and were followed by pigmentation and multifocal punctate fluorescein staining. Microscopically, findings were characterized by single-cell necrosis, pigmentation, disordered basilar layer, and thinning of the corneal epithelium. Increased toxicity was associated with a higher DAR or more stably attached linker. Treatment with agents to block target engagement did not affect toxicity, and none of the topical treatments was successful. Conclusions: The corneal findings observed were similar to the effects described in clinical trials with Depatux-m and other ADCs. Collectively, these studies and available literature support the hypothesis that ADC-mediated toxicity is driven primarily by mechanism of action of the payload.


Immunoconjugates , Toxic Optic Neuropathy , Animals , Antibodies, Monoclonal, Humanized/toxicity , Immunoconjugates/toxicity , Mice
5.
Blood ; 139(11): 1743-1759, 2022 03 17.
Article En | MEDLINE | ID: mdl-34986233

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative treatment of patients with nonmalignant or malignant blood disorders. Its success has been limited by graft-versus-host disease (GVHD). Current systemic nontargeted conditioning regimens mediate tissue injury and potentially incite and amplify GVHD, limiting the use of this potentially curative treatment beyond malignant disorders. Minimizing systemic nontargeted conditioning while achieving alloengraftment without global immune suppression is highly desirable. Antibody-drug-conjugates (ADCs) targeting hematopoietic cells can specifically deplete host stem and immune cells and enable alloengraftment. We report an anti-mouse CD45-targeted-ADC (CD45-ADC) that facilitates stable murine multilineage donor cell engraftment. Conditioning with CD45-ADC (3 mg/kg) was effective as a single agent in both congenic and minor-mismatch transplant models resulting in full donor chimerism comparable to lethal total body irradiation (TBI). In an MHC-disparate allo-HSCT model, pretransplant CD45-ADC (3 mg/kg) combined with low-dose TBI (150 cGy) and a short course of costimulatory blockade with anti-CD40 ligand antibody enabled 89% of recipients to achieve stable alloengraftment (mean value: 72%). When CD45-ADC was combined with pretransplant TBI (50 cGy) and posttransplant rapamycin, cyclophosphamide (Cytoxan), or a JAK inhibitor, 90% to 100% of recipients achieved stable chimerism (mean: 77%, 59%, 78%, respectively). At a higher dose (5 mg/kg), CD45-ADC as a single agent was sufficient for rapid, high-level multilineage chimerism sustained through the 22 weeks observation period. Therefore, CD45-ADC has the potential utility to confer the benefit of fully myeloablative conditioning but with substantially reduced toxicity when given as a single agent or at lower doses in conjunction with reduced-intensity conditioning.


Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Immunoconjugates , Animals , Chimerism , Hematopoietic Stem Cell Transplantation/adverse effects , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Immunoconjugates/toxicity , Mice , Transplantation Conditioning/methods
6.
Mol Pharm ; 18(11): 4058-4066, 2021 11 01.
Article En | MEDLINE | ID: mdl-34579528

To overcome a lack of selectivity during the chemical modification of native non-engineered antibodies, we have developed a technology platform termed "AJICAP" for the site-specific chemical conjugation of antibodies through the use of a class of IgG Fc-affinity reagents. To date, a limited number of antibody-drug conjugates (ADCs) have been synthesized via this approach, and no toxicological study was reported. Herein, we describe the compatibility and robustness of AJICAP technology, which enabled the synthesis of a wide variety of ADCs. A stability assessment of a thiol-modified antibody synthesized by AJICAP technology indicated no appreciable increase in aggregation or decomposition upon prolonged storage, indicating that the unexpectedly stable thiol intermediate has a great potential intermediate for payload or linker screening or large-scale manufacturing. Payload conjugation with this stable thiol intermediate generated several AJICAP-ADCs. In vivo xenograft studies indicated that the AJICAP-ADCs displayed significant tumor inhibition comparable to benchmark ADC Kadcyla. Furthermore, a rat pharmacokinetic analysis and toxicology study indicated an increase in the maximum tolerated dose, demonstrating an expansion of the AJICAP-ADC therapeutic index, compared with stochastic conjugation technology. This is the first report of the therapeutic index estimation of site-specific ADCs produced by utilizing Fc affinity reagent conjugation. The described site-specific conjugation technology is a powerful platform to enable next-generation ADCs through reduced heterogeneity and enhanced therapeutic index.


Antineoplastic Agents/pharmacokinetics , Drug Compounding/methods , Immunoconjugates/pharmacokinetics , Neoplasms/drug therapy , Ado-Trastuzumab Emtansine/administration & dosage , Ado-Trastuzumab Emtansine/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Chemistry, Pharmaceutical , Drug Stability , Female , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Immunoconjugates/toxicity , Maximum Tolerated Dose , Mice , Neoplasms/pathology , Rats , Therapeutic Index , Toxicity Tests, Acute , Xenograft Model Antitumor Assays
7.
Nat Commun ; 12(1): 3528, 2021 06 10.
Article En | MEDLINE | ID: mdl-34112795

Breast tumors generally consist of a diverse population of cells with varying gene expression profiles. Breast tumor heterogeneity is a major factor contributing to drug resistance, recurrence, and metastasis after chemotherapy. Antibody-drug conjugates (ADCs) are emerging chemotherapeutic agents with striking clinical success, including T-DM1 for HER2-positive breast cancer. However, these ADCs often suffer from issues associated with intratumor heterogeneity. Here, we show that homogeneous ADCs containing two distinct payloads are a promising drug class for addressing this clinical challenge. Our conjugates show HER2-specific cell killing potency, desirable pharmacokinetic profiles, minimal inflammatory response, and marginal toxicity at therapeutic doses. Notably, a dual-drug ADC exerts greater treatment effect and survival benefit than does co-administration of two single-drug variants in xenograft mouse models representing intratumor HER2 heterogeneity and elevated drug resistance. Our findings highlight the therapeutic potential of the dual-drug ADC format for treating refractory breast cancer and perhaps other cancers.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Immunoconjugates/pharmacology , Receptor, ErbB-2/immunology , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/complications , Cell Line, Tumor , Female , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Immunoconjugates/toxicity , Immunohistochemistry , Inflammation/complications , Mice , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Xenograft Model Antitumor Assays
8.
MAbs ; 13(1): 1914885, 2021.
Article En | MEDLINE | ID: mdl-33904380

Antibody-drug conjugates (ADCs) are targeted therapeutic agents that treat cancers by selective delivery of highly potent cytotoxic drugs to tumor cells via cancer-specific antibodies. However, their clinical benefit is limited by off-target toxicity and narrow therapeutic windows. To overcome these limitations, we have applied reductive alkylation to develop a new type of ADC that has cytotoxic drugs conjugated to the N-terminal of an antibody through amine bonds introduced via reductive alkylation reactions (NTERM). To test whether the NTERM-conjugated ADCs can widen therapeutic windows, we synthesized three different ADCs by conjugating trastuzumab and monomethyl auristatin-F using three different methods, and compared their stability, efficacy, and toxicity. The NTERM-conjugated ADC was more stable in vitro and in vivo than the thiol-conjugated and the lysine-conjugated ADCs. The NTERM-conjugated ADC showed lower toxicity compared to other ADCs, whereas its efficacy was comparable to that of the thiol-conjugated ADC and better than that of the lysine-conjugated ADC. These results suggest that the NTERM conjugation method could widen the therapeutic window of ADCs by enhancing its stability and reducing toxicity.


Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/drug therapy , Immunoconjugates/pharmacology , Oligopeptides/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Trastuzumab/pharmacology , Alkylation , Animals , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/toxicity , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Compounding , Drug Stability , Female , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/toxicity , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Oligopeptides/toxicity , Protein Stability , Rats, Nude , Rats, Sprague-Dawley , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Trastuzumab/chemistry , Trastuzumab/pharmacokinetics , Trastuzumab/toxicity , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Bioconjug Chem ; 32(4): 746-754, 2021 04 21.
Article En | MEDLINE | ID: mdl-33689309

Although peptide motifs represent the majority of cleavable linkers used in clinical-stage antibody-drug conjugates (ADCs), the sequences are often sensitive to cleavage by extracellular enzymes, such as elastase, which leads to systemic release of the cytotoxic payload. This action reduces the therapeutic index by causing off-target toxicities that can be dose-limiting. For example, a common side-effect of ADCs made using peptide-cleavable linkers is myelosuppression, including neutropenia. Only a few reports describe methods for optimizing peptide linkers to maintain efficient and potent tumor payload delivery while enhancing circulating stability. Herein, we address these critical limitations through the development of a tandem-cleavage linker strategy, where two sequential enzymatic cleavage events mediate payload release. We prepared dipeptides that are protected from degradation in the circulation by a sterically encumbering glucuronide moiety. Upon ADC internalization and lysosomal degradation, the monosaccharide is removed and the exposed dipeptide is degraded, which liberates the attached payload inside the target cell. We used CD79b-targeted monomethyl auristatin E (MMAE) conjugates as our model system and compared the stability, efficacy, and tolerability of ADCs made with tandem-cleavage linkers to ADCs made using standard technology with the vedotin linker. The results, where rat studies showed dramatically improved tolerability in the hematopoietic compartment, highlight the role that linker stability plays in efficacy and tolerability and also offer a means of improving an ADC's therapeutic index for improved patient outcomes.


Antineoplastic Agents/toxicity , CD79 Antigens/toxicity , Immunoconjugates/toxicity , Animals , Antineoplastic Agents/chemistry , CD79 Antigens/chemistry , Endocytosis , Female , Hydrolysis , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , In Vitro Techniques , Male , Mice , Mice, Inbred NOD , Mice, SCID , Rats , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays
10.
BMC Cancer ; 21(1): 270, 2021 Mar 12.
Article En | MEDLINE | ID: mdl-33711962

BACKGROUND: Epidermal growth factor receptor (EGFR) is a target for cancer therapy as it is overexpressed in a wide variety of cancers. Therapeutic antibodies that bind EGFR are being evaluated in clinical trials as imaging agents for positron emission tomography and image-guided surgery. However, some of these antibodies have safety concerns such as infusion reactions, limiting their use in imaging applications. Nimotuzumab is a therapeutic monoclonal antibody that is specific for EGFR and has been used as a therapy in a number of countries. METHODS: Formulation of IRDye800CW-nimotuzumab for a clinical trial application was prepared. The physical, chemical, and pharmaceutical properties were tested to develop the specifications to determine stability of the product. The acute and delayed toxicities were tested and IRDye800CW-nimotuzumab was determined to be non-toxic. Non-compartmental pharmacokinetics analysis was used to determine the half-life of IRDye800CW-nimotuzumab. RESULTS: IRDye800CW-nimotuzumab was determined to be non-toxic from the acute and delayed toxicity study. The half-life of IRDye800CW-nimotuzumab was determined to be 38 ± 1.5 h. A bi-exponential analysis was also used which gave a t1/2 alpha of 1.5 h and t1/2 beta of 40.8 h. CONCLUSIONS: Here, we show preclinical studies demonstrating that nimotuzumab conjugated to IRDye800CW is safe and does not exhibit toxicities commonly associated with EGFR targeting antibodies.


Drugs, Investigational/administration & dosage , Immunoconjugates/administration & dosage , Neoplasms/diagnostic imaging , Optical Imaging/methods , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/toxicity , Benzenesulfonates/administration & dosage , Benzenesulfonates/pharmacokinetics , Benzenesulfonates/toxicity , Cell Line, Tumor , Clinical Trials as Topic , Drug Stability , Drugs, Investigational/pharmacology , Drugs, Investigational/toxicity , ErbB Receptors/antagonists & inhibitors , Female , Half-Life , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/toxicity , Indoles/administration & dosage , Indoles/pharmacokinetics , Indoles/toxicity , Investigational New Drug Application , Male , Mice , Neoplasms/pathology , Neoplasms/surgery , Surgery, Computer-Assisted/methods , Toxicity Tests, Acute , Xenograft Model Antitumor Assays
11.
J Immunol Methods ; 483: 112794, 2020 08.
Article En | MEDLINE | ID: mdl-32428450

A commonly employed method to determine the function of a particular cell population and to assess its contribution to the overall system in vivo is to selectively deplete that population and observe the effects. Using monoclonal antibodies to deliver toxins to target cells can achieve this with a high degree of efficiency. Here, we describe an in vivo model combining the use of immunotoxins and multidrug resistant (MDR) gene deficient mice so that only MDR deficient cells expressing the target molecule would be depleted while target molecule expressing, but MDR sufficient, cells are spared. This allows targeted depletion at a higher degree of specificity than has been previously achieved. We have applied this technique to study trogocytosis, the intercellular transfer of cell surface molecules, but this principle could also be adapted using technology already available for use in other fields of study.


ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cytotoxicity, Immunologic/drug effects , Genes, MDR/physiology , Immunotoxins/toxicity , Lymphocyte Depletion/methods , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/deficiency , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Animals , Female , Graft Survival/drug effects , Heart Transplantation , Histocompatibility Antigens Class II/immunology , Immunoconjugates/toxicity , Immunoglobulin Fab Fragments/toxicity , Kidney Transplantation , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Ribosome Inactivating Proteins, Type 1/toxicity , Spleen/drug effects , Spleen/immunology , Spleen/pathology , Transplantation Tolerance/drug effects
12.
ACS Appl Mater Interfaces ; 12(21): 23717-23725, 2020 May 27.
Article En | MEDLINE | ID: mdl-32368886

Antibody-drug conjugates have shown great promise in active targeting for cancer therapy. The existing chemical techniques for antibody conjugation generally lack efficiency or universality. In this article, a site-specific antibody conjugation was developed by using a mild reaction between a benzoboroxole (BB) functionality and cis-diol moiety of sugar units in the antibody fragment crystallizable region under neutral pH conditions. A BB/PEG/ICG-grafted poly(aspartic acid) comb-like functional polymer was first synthesized and conjugated with transferrin (Tf) to form a transferrin-polymer-drug conjugate [Tf-P(BB)], which showed 120% increase in HepG2 hepatoma (Tf receptor overexpression) cell uptake compared to a nontargeting protein-polymer-drug conjugate [HRP-P(BB)]. The universality of this method was further demonstrated by the enhanced uptake of trastuzumab (anti-Her2 antibody)-polymer-drug conjugates in MCF-7 (295%) and MDA-MB-435S (66.4%) (Her2 positive) cells. The positive charge of the linker had great influence on the targeting ability of the antibody-polymer-drug conjugates. The in vivo studies demonstrated the distinct targeting ability of Tf-P(BB) in the HepG2 xenograft tumor, and the tumor accumulation of the Tf-P(BB) testing group increased by 92% with respect to the control group [HRP-P(BB)]. More significantly, the HepG2 cell uptake amount of the antibody-oriented conjugate [Tf-P'(BB)] was 2.4-fold higher than that of the controlled group [Tf-P'(Hex)]. On the basis of this facile site-specific conjugation method, the conjugates are able to change the antibody species easily against various cancers, while maintaining the antibody integrity and targeting ability.


Antibodies/chemistry , Immunoconjugates/chemistry , Neoplasms/immunology , Animals , Antibodies/immunology , Antibodies/toxicity , Boron Compounds/chemistry , Boron Compounds/toxicity , Cell Line, Tumor , Female , HEK293 Cells , Humans , Immunoconjugates/immunology , Immunoconjugates/toxicity , Mice, Inbred BALB C , Peptides/chemical synthesis , Peptides/toxicity , Transferrin/chemistry , Transferrin/toxicity
13.
Eur J Med Chem ; 190: 112080, 2020 Mar 15.
Article En | MEDLINE | ID: mdl-32018094

Disulfide re-bridging strategy has demonstrated significant advantages in the construction of homogeneous antibody drug conjugates (ADCs). However, a major issue that disulfide scrambling at the hinge region of antibody leads to the formation of "half-antibody" has appeared for many re-bridging linkers. We present bis(vinylsulfonyl)piperazines (BVP) as efficient linkers to selectively re-bridge disulfides at the antigen-binding fragment (Fab) regions and produce highly homogeneous conjugates with a loading of two drugs without disulfide scrambling. We also found that optically active (S)-configuration linkers led to more sufficient conjugation compared with (R)-configuration. The BVP-linked ADCs demonstrated superior efficacy and antigen-selectivity in vitro cytotoxicity.


Immunoconjugates/pharmacology , Piperazines/pharmacology , Sulfones/pharmacology , Vinyl Compounds/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Cell Line, Tumor , Disulfides/chemistry , Drug Screening Assays, Antitumor , Humans , Immunoconjugates/chemistry , Immunoconjugates/toxicity , Piperazines/chemical synthesis , Piperazines/toxicity , Sulfones/chemical synthesis , Sulfones/toxicity , Vinyl Compounds/chemical synthesis , Vinyl Compounds/toxicity
14.
Mol Oncol ; 14(1): 54-68, 2020 01.
Article En | MEDLINE | ID: mdl-31736230

cMet is a well-characterized oncogene that is the target of many drugs including small molecule and biologic pathway inhibitors, and, more recently, antibody-drug conjugates (ADCs). However, the clinical benefit from cMet-targeted therapy has been limited. We developed a novel cMet-targeted 'third-generation' ADC, TR1801-ADC, that was optimized at different levels including specificity, stability, toxin-linker, conjugation site, and in vivo efficacy. Our nonagonistic cMet antibody was site-specifically conjugated to the pyrrolobenzodiazepine (PBD) toxin-linker tesirine and has picomolar activity in cancer cell lines derived from different solid tumors including lung, colorectal, and gastric cancers. The potency of our cMet ADC is independent of MET gene copy number, and its antitumor activity was high not only in high cMet-expressing cell lines but also in medium-to-low cMet cell lines (40 000-90 000 cMet/cell) in which a cMet ADC with tubulin inhibitor payload was considerably less potent. In vivo xenografts with low-medium cMet expression were also very responsive to TR1801-ADC at a single dose, while a cMet ADC using a tubulin inhibitor showed a substantially reduced efficacy. Furthermore, TR1801-ADC had excellent efficacy with significant antitumor activity in 90% of tested patient-derived xenograft models of gastric, colorectal, and head and neck cancers: 7 of 10 gastric models, 4 of 10 colorectal cancer models, and 3 of 10 head and neck cancer models showed complete tumor regression after a single-dose administration. Altogether, TR1801-ADC is a new generation cMet ADC with best-in-class preclinical efficacy and good tolerability in rats.


Antineoplastic Agents/pharmacology , Benzodiazepines/pharmacology , Immunoconjugates/pharmacology , Neoplasms/drug therapy , Oncogenes/immunology , Proto-Oncogene Proteins c-met/immunology , Pyrroles/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Biliary Tract Neoplasms/metabolism , Cell Line, Tumor , Colonic Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/metabolism , Humans , Immunoconjugates/therapeutic use , Immunoconjugates/toxicity , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/immunology , Proto-Oncogene Proteins c-met/metabolism , Rats , Rats, Sprague-Dawley , Stomach Neoplasms/metabolism , Tissue Array Analysis , Xenograft Model Antitumor Assays
15.
J Pharm Sci ; 109(1): 761-768, 2020 01.
Article En | MEDLINE | ID: mdl-31376374

Closed system transfer devices (CSTD) are a supplemental engineering control designed to reduce occupational exposure of hazardous drugs and are currently implemented in accordance with evolving regulations. Owing to the novelty and complexity of these devices and their importance in clinical in-use testing, here we evaluated FDA-approved CSTD, assessing product quality through stability indicating assays to determine any drug product incompatibilities. Six devices were used in a simulated compounding and administration of a late-phase IgG1 antibody-drug conjugate (ADC) and the resulting samples were analyzed for visible and subvisible particle counts by light obscuration and micro-flow imaging, physical stability by size exclusion chromatography, and biological activities by relative potency. Potential challenges included improper fit of CSTD components, loss of product to void volume, and material incompatibility. Results showed compatibility of the ADC with the 6 CSTD evaluated. One CSTD introduced subvisible particles into the ADC during compounding that were identified through morphological assessment as silicone oil. This study highlights the importance of clinical in use testing with new devices and proposes strategies to mitigate the risk of drug product incompatibility with CSTD.


Drug Compounding/instrumentation , Immunoconjugates/chemistry , Immunoglobulin G/chemistry , Occupational Exposure/prevention & control , Protective Devices , Cell Line, Tumor , Cell Survival/drug effects , Drug Stability , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/toxicity , Immunoglobulin G/administration & dosage , Immunoglobulin G/toxicity , Materials Testing , Occupational Exposure/adverse effects , Protein Aggregates , Protein Stability
16.
Toxicol Lett ; 324: 30-37, 2020 May 15.
Article En | MEDLINE | ID: mdl-31877330

The HER2 pathway plays a pivotal role in cell proliferation and differentiation, while the receptor overexpression caused by amplification of HER2 gene is associated with the growth of several tumors. Previously published clinical trials have demonstrated that antibody-conjugated drugs (ADCs) remarkably improved clinical effects compared with antibodies alone for the same target. In order to provide more effective drugs, we developed Disitamab vedotin based on ADC. The antibody part was a humanized monoclonal antibody targeting HER2, the small molecule toxin was monomethyl auristatin E (MMAE), a synthetic antineoplastic agent. A protease cleavable linker covalently attached MMAE to the antibody. In this study, we characterized the toxicity profile of Disitamab vedotin through single- and repeat-dose toxicity studies in monkeys. The toxicities of small molecules and naked antibody (Disitamab) were also assessed in these studies. Monkeys were well tolerated with Disitamab vedotin at doses of 6 mg/kg, while equivalent MMAEs resulted in severe myelosuppression. This finding proves that ADCs improve the therapeutic effect. In addition, the safety profiles of Disitamab vedotin and MMAE were similar and consistent with the activation mechanism of MMAE. Toxicology finding included bone marrow/hematology toxicity and lymphoid organ toxicity, while no significant toxicity was observed in animals treated with naked antibody. These side effects were found to be consistent with data acquired from clinical phase I/II patients treated with Disitamab vedotin.


Antineoplastic Agents/toxicity , Immunoconjugates/toxicity , Oligopeptides/toxicity , Receptor, ErbB-2/antagonists & inhibitors , Animals , Cross Reactions , Immunoconjugates/pharmacokinetics , Macaca fascicularis , Oligopeptides/pharmacokinetics , Rats , Rats, Sprague-Dawley
17.
ACS Chem Biol ; 14(10): 2233-2242, 2019 10 18.
Article En | MEDLINE | ID: mdl-31513374

Recent studies have shown that modified human lactoferrin 20-31 fragment, named HLopt2, possesses antibacterial and antifungal activity. Thus, we decided to synthesize and evaluate the biological activity of a series of conjugates based on this peptide and one of the antimicrobials with proven antibacterial (ciprofloxacin, CIP, and levofloxacin, LVX) or antifungal (fluconazole, FLC) activity. The drugs were covalently connected to the peptide via amide, methylenecarbonyl moieties, or a disulfide bridge. The antibacterial and antifungal activities were evaluated under Clinical and Laboratory Standard Institute (CLSI) recommended conditions or in a low-salt brain-heart infusion diluted medium (BHI1/100). Results showed that conjugation of the peptide with the drug increased its antimicrobial activity up to 4-fold. Under CLSI-recommended conditions, all the compounds revealed rather low efficiency. Among conjugates, the highest antibacterial activity was recorded for the CIP-Cys-S-S-HLopt2-NH2 (III). In BHI1/100, which had lower differentiating properties, all of the conjugates revealed low MIC and MMC (minimum inhibitory and microbicidal concentrations) values. The disulfide bridge used as a linker in the most active conjugate (III) upon incubation with S. aureus cells is reduced, releasing constituent peptide and CIP-Cys. In addition, we showed that its fluorescently labeled analogue and constituent peptide are able to be internalized into both C. albicans and S. aureus cells. Moreover, the invaluable advantage of the presented conjugates was their low toxicity to mammalian cells and very low hemolytic activity. The current research can form a solid basis for further in vivo studies and drug development.


Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Immunoconjugates/pharmacology , Lactoferrin/pharmacology , Peptide Fragments/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/toxicity , Antifungal Agents/chemical synthesis , Antifungal Agents/toxicity , Candida albicans/drug effects , Ciprofloxacin/chemical synthesis , Ciprofloxacin/pharmacology , Ciprofloxacin/toxicity , Drug Stability , Escherichia coli/drug effects , Fluconazole/chemical synthesis , Fluconazole/pharmacology , Fluconazole/toxicity , HEK293 Cells , Hep G2 Cells , Humans , Immunoconjugates/toxicity , Lactoferrin/chemical synthesis , Lactoferrin/toxicity , Levofloxacin/chemical synthesis , Levofloxacin/pharmacology , Levofloxacin/toxicity , Male , Microbial Sensitivity Tests , Peptide Fragments/chemical synthesis , Peptide Fragments/toxicity , Staphylococcus aureus/drug effects , Swine
18.
Nat Chem Biol ; 15(10): 949-958, 2019 10.
Article En | MEDLINE | ID: mdl-31451760

Antibody-drug conjugates (ADCs) selectively deliver chemotherapeutic agents to target cells and are important cancer therapeutics. However, the mechanisms by which ADCs are internalized and activated remain unclear. Using CRISPR-Cas9 screens, we uncover many known and novel endolysosomal regulators as modulators of ADC toxicity. We identify and characterize C18ORF8/RMC1 as a regulator of ADC toxicity through its role in endosomal maturation. Through comparative analysis of screens with ADCs bearing different linkers, we show that a subset of late endolysosomal regulators selectively influence toxicity of noncleavable linker ADCs. Surprisingly, we find cleavable valine-citrulline linkers can be processed rapidly after internalization without lysosomal delivery. Lastly, we show that sialic acid depletion enhances ADC lysosomal delivery and killing in diverse cancer cell types, including with FDA (US Food and Drug Administration)-approved trastuzumab emtansine (T-DM1) in Her2-positive breast cancer cells. Together, these results reveal new regulators of endolysosomal trafficking, provide important insights for ADC design and identify candidate combination therapy targets.


CRISPR-Cas Systems , Genome-Wide Association Study , Immunoconjugates/toxicity , Maytansine/analogs & derivatives , N-Acetylneuraminic Acid/pharmacology , Trastuzumab/pharmacology , Ado-Trastuzumab Emtansine , Antineoplastic Agents, Immunological/pharmacology , Carrier Proteins , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Lysosomes , Maytansine/pharmacology
19.
Regul Toxicol Pharmacol ; 107: 104429, 2019 Oct.
Article En | MEDLINE | ID: mdl-31325532

With a new generation of antibody-drug conjugates (ADCs) that contain a drug-to-antibody ratio (DAR) of 2, the question remains whether advances in technology have resulted in more stable and tumor-specific ADCs. These ADCs are anticipated to cause minimal systemic exposures of payloads, with toxicities being evident mainly at tumor sites. We examined 15 ADCs with PBD-dimer payloads and a DAR of 2 and concluded that dose limiting toxicities in animals and in humans are generally related to the payload. Both the payloads and the ADCs had pro-inflammatory responses causing severe toxicities that were at times of low incidence, making it difficult to assess a cause-effect relationship. Due to their low incidence, single-patient cohorts may not detect these events and such design may not be suitable in first-in-human (FIH) trials. The commonly proposed approach by the sponsors for FIH dose selection was 1/6th highest non-severely toxic dose (HNSTD) in monkeys. This approach resulted in an acceptable balance of safety and efficient dose escalation in phase 1 trials, when using data from repeat-dose toxicology studies and body surface area for scaling. No sponsor used the data generated in rodents or proposed novel approaches for FIH dose selection.


Benzodiazepines/toxicity , Immunoconjugates/toxicity , Pyrroles/toxicity , Animals , Haplorhini , Humans , Maximum Tolerated Dose , Mice , Rats
20.
Chembiochem ; 20(18): 2411-2419, 2019 09 16.
Article En | MEDLINE | ID: mdl-31044494

Microbial transglutaminase (mTG) has recently emerged as a powerful tool for antibody engineering. In nature, it catalyzes the formation of amide bonds between glutamine side chains and primary amines. Being applied to numerous research fields from material sciences to medicine, mTG enables efficient site-specific conjugation of molecular architectures that possess suitable recognition motifs. In monoclonal antibodies, the lack of native transamidation sites is bypassed by incorporating specific peptide recognition sequences. Herein, we report a rapid and efficient mTG-catalyzed bioconjugation that relies on a novel recognition motif derived from its native substrate Streptomyces papain inhibitor (SPIP ). Improved reaction kinetics compared to commonly applied sequences were demonstrated for model peptides and for biotinylation of Her2-targeting antibody trastuzumab variants. Moreover, an antibody-drug conjugate assembled from trastuzumab that was C-terminally tagged with the novel recognition sequence revealed a higher payload-antibody ratio than the reference antibody.


Antineoplastic Agents, Immunological/chemistry , Bacterial Proteins/chemistry , Immunoconjugates/chemistry , Oligopeptides/chemistry , Transglutaminases/chemistry , Trastuzumab/chemistry , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/toxicity , CHO Cells , Cell Line, Tumor , Cricetulus , Humans , Immunoconjugates/toxicity , Oligopeptides/pharmacology , Oligopeptides/toxicity , Protein Engineering , Streptomyces/enzymology , Substrate Specificity , Trastuzumab/pharmacology , Trastuzumab/toxicity
...