Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Rev Clin Oncol ; 21(3): 203-223, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38191923

ABSTRACT

Antibody-drug conjugates (ADCs) are a promising cancer treatment modality that enables the selective delivery of highly cytotoxic payloads to tumours. However, realizing the full potential of this platform necessitates innovative molecular designs to tackle several clinical challenges such as drug resistance, tumour heterogeneity and treatment-related adverse effects. Several emerging ADC formats exist, including bispecific ADCs, conditionally active ADCs (also known as probody-drug conjugates), immune-stimulating ADCs, protein-degrader ADCs and dual-drug ADCs, and each offers unique capabilities for tackling these various challenges. For example, probody-drug conjugates can enhance tumour specificity, whereas bispecific ADCs and dual-drug ADCs can address resistance and heterogeneity with enhanced activity. The incorporation of immune-stimulating and protein-degrader ADCs, which have distinct mechanisms of action, into existing treatment strategies could enable multimodal cancer treatment. Despite the promising outlook, the importance of patient stratification and biomarker identification cannot be overstated for these emerging ADCs, as these factors are crucial to identify patients who are most likely to derive benefit. As we continue to deepen our understanding of tumour biology and refine ADC design, we will edge closer to developing truly effective and safe ADCs for patients with treatment-refractory cancers. In this Review, we highlight advances in each ADC component (the monoclonal antibody, payload, linker and conjugation chemistry) and provide more-detailed discussions on selected examples of emerging novel ADCs of each format, enabled by engineering of one or more of these components.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Neoplasms , Humans , Immunoconjugates/chemistry , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/therapeutic use , Pharmaceutical Preparations
2.
Mol Cancer Ther ; 21(9): 1449-1461, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35793453

ABSTRACT

Valine-citrulline is a protease-cleavable linker commonly used in many drug delivery systems, including antibody-drug conjugates (ADC) for cancer therapy. However, its suboptimal in vivo stability can cause various adverse effects such as neutropenia and hepatotoxicity, leading to dose delays or treatment discontinuation. Here, we report that glutamic acid-glycine-citrulline (EGCit) linkers have the potential to solve this clinical issue without compromising the ability of traceless drug release and ADC therapeutic efficacy. We demonstrate that our EGCit ADC resists neutrophil protease-mediated degradation and spares differentiating human neutrophils. Notably, our anti-HER2 ADC shows almost no sign of blood and liver toxicity in healthy mice at 80 mg kg-1. In contrast, at the same dose level, the FDA-approved anti-HER2 ADCs Kadcyla and Enhertu show increased levels of serum alanine aminotransferase and aspartate aminotransferase and morphologic changes in liver tissues. Our EGCit conjugates also exert greater antitumor efficacy in multiple xenograft tumor models compared with Kadcyla and Enhertu. This linker technology could substantially broaden the therapeutic windows of ADCs and other drug delivery agents, providing clinical options with improved efficacy and safety.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Ado-Trastuzumab Emtansine , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Citrulline , Humans , Immunoconjugates/metabolism , Immunoconjugates/pharmacology , Mice , Peptide Hydrolases , Therapeutic Index
3.
RSC Adv ; 12(6): 3359-3364, 2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35425350

ABSTRACT

Antibody-based therapy has shown great success in the treatment of many diseases, including cancers. While antibodies and antibody-drug conjugates (ADCs) have also been evaluated for central nervous system (CNS) disorders as well as brain tumors, their therapeutic efficacy can be substantially limited due to low permeability across the blood-brain barrier (BBB). Thus, improving BBB permeability of therapeutic antibodies is critical in establishing this drug class as a reliable clinical option for CNS diseases. Here, we report that, compared with a conventional heterogeneous conjugation, homogeneous conjugation of the synthetic BBB shuttle peptide angiopep-2 (Ang2) to a monoclonal antibody (mAb) provides improved binding affinity for brain microvascular endothelial cells in vitro and accumulation into normal brain tissues in vivo. In a mouse model, we also demonstrate that the homogeneous anti-EGFR mAb-Ang2 conjugate administered intravenously efficiently accumulates in intracranial tumors. These findings suggest that homogeneous conjugation of BBB shuttle peptides such as Ang2 is a promising approach to enhancing the therapeutic efficacy of antibody agents for CNS diseases.

4.
Nat Commun ; 12(1): 3528, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112795

ABSTRACT

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.


Subject(s)
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
5.
Mol Cancer Ther ; 19(11): 2330-2339, 2020 11.
Article in English | MEDLINE | ID: mdl-32879051

ABSTRACT

Acute myeloid leukemia (AML) is the most common and aggressive blood cancer in adults. In particular, significant unmet medical needs exist for effective treatment strategies for acute myelomonocytic leukemia (M4) and acute monocytic leukemia (M5) AML subtypes. Antibody-drug conjugates (ADC) are a promising drug class for AML therapy, as demonstrated by the FDA-approved anti-CD33 ADC, gemtuzumab ozogamicin (Mylotarg). However, CD33 is expressed in normal hematopoietic stem cells, highlighting the critical need to identify AML-specific targets to minimize the risk of potential adverse effects. We have demonstrated that the leukocyte immunoglobulin-like receptor subfamily B4 (LILRB4) is expressed at significantly higher levels on monocytic M4 and M5 AML cells than on normal counterparts. Here, we test whether LILRB4 is a promising ADC target to kill monocytic AML cells while sparing healthy counterparts. To this end, we generated ADCs from a humanized anti-LILRB4 mAb and the antimitotic payload, monomethyl auristatin F. The conjugates constructed were characterized and evaluated for LILRB4-specific cell killing potency, toxicity to progenitor cells, pharmacokinetics, and therapeutic efficacy. Our ADC linker technology platform efficiently generated homogeneous anti-LILRB4 ADCs with defined drug-to-antibody ratios. The homogeneous anti-LILRB4 ADCs demonstrated the capacity for LILRB4-mediated internalization, suitable physicochemical properties, and high cell killing potency against LILRB4-positive AML cells. Importantly, our data indicate that these ADCs spare normal progenitor cells. One of our homogeneous conjugates exerted a remarkable therapeutic effect and no significant toxicity in a xenograft mouse model of disseminated human AML. Our findings highlight the clinical potential of anti-LILRB4 ADCs in monocytic AML therapy.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Immunoconjugates/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Receptors, Immunologic/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/chemistry , Cell Line, Tumor , Disease Models, Animal , Drug Stability , Humans , Hydrophobic and Hydrophilic Interactions , Immunoconjugates/chemistry , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Mice , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Nat Commun ; 9(1): 2512, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29955061

ABSTRACT

Valine-citrulline linkers are commonly used as enzymatically cleavable linkers for antibody-drug conjugates. While stable in human plasma, these linkers are unstable in mouse plasma due to susceptibility to an extracellular carboxylesterase. This instability often triggers premature release of drugs in mouse circulation, presenting a molecular design challenge. Here, we report that an antibody-drug conjugate with glutamic acid-valine-citrulline linkers is responsive to enzymatic drug release but undergoes almost no premature cleavage in mice. We demonstrate that this construct exhibits greater treatment efficacy in mouse tumor models than does a valine-citrulline-based variant. Notably, our antibody-drug conjugate contains long spacers facilitating the protease access to the linker moiety, indicating that our linker assures high in vivo stability despite a high degree of exposure. This technology could add flexibility to antibody-drug conjugate design and help minimize failure rates in pre-clinical studies caused by linker instability.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Glutamic Acid/chemistry , Immunoconjugates/pharmacology , Oligopeptides/chemistry , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carboxylic Ester Hydrolases/blood , Cell Line, Tumor , Citrulline/chemistry , Drug Design , Drug Stability , Female , Gene Expression , Humans , Hydrolysis , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Mice , Mice, Inbred BALB C , Mice, Nude , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Survival Analysis , Valine/chemistry , Xenograft Model Antitumor Assays
7.
J Control Release ; 212: 59-69, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26071630

ABSTRACT

Peritoneal carcinomatosis is a major source of morbidity and mortality in patients with advanced abdominal neoplasms. Intraperitoneal chemotherapy (IPC) is an area of intense interest given its efficacy in ovarian cancer. However, IPC suffers from poor drug penetration into peritoneal tumors. As such, extensive cytoreductive surgery is required prior to IPC. Here, we explore the utility of iRGD, a tumor-penetrating peptide, for improved tumor-specific penetration of intraperitoneal compounds and enhanced IPC in mice. Intraperitoneally administered iRGD significantly enhanced penetration of an attached fluorescein into disseminated peritoneal tumor nodules. The penetration was tumor-specific, circulation-independent, and mediated by the neuropilin-binding RXXK tissue-penetration peptide motif of iRGD. Q-iRGD, which fluoresces upon cleavage, including the one that leads to RXXK activation, specifically labeled peritoneal metastases displaying different growth patterns in mice. Importantly, iRGD enhanced intratumoral entry of intraperitoneally co-injected dextran to approximately 300% and doxorubicin to 250%. Intraperitoneal iRGD/doxorubicin combination therapy inhibited the growth of bulky peritoneal tumors and reduced systemic drug toxicity. iRGD delivered attached fluorescein and co-applied nanoparticles deep into fresh human peritoneal metastasis explants. These results indicate that intraperitoneal iRGD co-administration serves as a simple and effective strategy to facilitate tumor detection and improve the therapeutic index of IPC for peritoneal carcinomatosis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma/drug therapy , Cell-Penetrating Peptides/administration & dosage , Doxorubicin/administration & dosage , Oligopeptides/administration & dosage , Peritoneal Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell-Penetrating Peptides/therapeutic use , Doxorubicin/therapeutic use , Humans , Mice, Nude , Oligopeptides/therapeutic use , Regional Blood Flow
9.
Biopolymers ; 100(6): 705-13, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23494659

ABSTRACT

Collagen-model peptides composed of (X-Y-Gly)n sequences were used to study the triple helical structure of collagen. We report the stability of these collagen-like peptides in biological fluids, and their pharmacokinetics including distribution, metabolism, and excretion in animals. A typical collagen-model peptide, H-(Pro-Hyp-Gly)10-OH, was found to be extremely stable in the plasma and distributed mainly in the vascular blood space, and was eliminated through glomerular filtration in the kidneys. Triple helical peptides of (X-Y-Gly)n sequences were quantitatively recovered from the urine of rats after intravenous injection regardless of the differences in peptide net charge between -3 and +6 per triple helix. In contrast, the renal clearance became less efficient when the number of triplet repeats (n) was 12 or more. We also demonstrated the application of a collagen-like triple helical peptide as a novel drug carrier in the blood with a high urinary excretion profile. We further demonstrated that a collagen-like triple helical peptide conjugated to a spin probe, PROXYL, has the potential to evaluate the redox status of oxidative stress-induced animals in vivo.


Subject(s)
Drug Carriers , Peptides , Animals , Collagen/chemistry , Peptides/chemistry , Protein Conformation , Protein Structure, Secondary
10.
Macromol Rapid Commun ; 33(10): 911-5, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22389192

ABSTRACT

Square-millimeter-sized free-floating translucent films are formed in physiological buffer by multiway connections between biotinylated collagen-like triple-helical peptides and avidin. Although the compositions of the films are almost constant, regardless of the ratios of the components loaded, their thicknesses can be controlled by the concentrations of the components. The film surfaces can be further modified by taking advantage of exposed biotin (or avidin) functionalities. The self-assembled films could serve as novel materials in biomedical and biosensing applications.


Subject(s)
Collagen/chemistry , Peptides/chemistry , Amino Acid Sequence , Avidin/chemistry , Fluorescein/chemistry , Peptides/chemical synthesis , Protein Structure, Secondary , Rhodamines/chemistry , Surface Properties
11.
J Biol Chem ; 286(30): 26364-74, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21652703

ABSTRACT

Pigment epithelium-derived factor (PEDF) is a collagen-binding protein that is abundantly distributed in various tissues, including the eye. It exhibits various biological functions, such as anti-angiogenic, neurotrophic, and neuroprotective activities. PEDF also interacts with extracellular matrix components such as collagen, heparan sulfate proteoglycans (HSPGs), and hyaluronan. The collagen-binding property has been elucidated to be important for the anti-angiogenic activity in vivo (Hosomichi, J., Yasui, N., Koide, T., Soma, K., and Morita, I. (2005) Biochem. Biophys. Res. Commun. 335, 756-761). Here, we investigated the collagen recognition mechanism by PEDF. We first narrowed down candidate PEDF-binding sequences by taking advantage of previously reported structural requirements in collagen. Subsequent searches for PEDF-binding sequences employing synthetic collagen-like peptides resulted in the identification of one of the critical binding sites for PEDF, human α1(I)(929-938) (IKGHRGFSGL). Further analysis revealed that the collagen recognition by PEDF is sequence- and conformation-specific, and the high affinity binding motif is KGXRGFXGL in the triple helix. The PEDF-binding motif significantly overlapped with the heparin/HSPG-binding motif, KGHRG(F/Y). The interaction of PEDF with collagen I was specifically competed with by heparin but not by chondroitin sulfate-C or hyaluronan. The binding sequences for PEDF and heparin/HSPG also overlapped with the covalent cross-linking sites between collagen molecules. These findings imply a functional relationship between PEDF and HSPGs during angiogenesis, and the interaction of these molecules is regulated by collagen modifications.


Subject(s)
Collagen Type I/chemistry , Eye Proteins/chemistry , Heparan Sulfate Proteoglycans/chemistry , Heparin/chemistry , Nerve Growth Factors/chemistry , Peptides/chemistry , Serpins/chemistry , Amino Acid Motifs , Animals , Binding Sites , Cattle , Collagen Type I/genetics , Collagen Type I/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Heparin/genetics , Heparin/metabolism , Humans , Mice , Neovascularization, Physiologic/physiology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Peptides/genetics , Peptides/metabolism , Serpins/genetics , Serpins/metabolism
12.
Biomaterials ; 31(7): 1925-34, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19853297

ABSTRACT

Collagen is an abundantly distributed extracellular matrix protein in mammalian bodies that maintains structural integrity of the organs and tissues. Besides its function as a structural protein, collagen has various biological functions which regulate cell adhesion, migration and differentiation. In order to develop totally synthetic collagen-surrogates, we recently reported a basic concept for preparing collagen-like triple helical supramolecules based on the self-assembly of staggered trimeric peptides with self-complementary shapes. In this paper, we add one of the specific cellular functions of the native collagen to the collagen-mimetic supramolecule. We synthesized a self-assembling peptide unit containing the integrin-binding sequence Gly-Phe-Hyp-Gly-Glu-Arg. The supramolecule carrying the sequence exhibited significant binding activity to human dermal fibroblasts. The supramolecular structure was found to be essential for function in in vitro cell culture. Cell adhesion was shown to be comparable to that of native collagen, and was further demonstrated to be mediated solely by integrin alpha 2 beta 1. Well-grown focal contacts and stress fibers were observed in cells spread on the supramolecular collagen-mimetic. The results demonstrate the potential of peptide-based artificial collagen as a biomaterial for regulating specific cellular function and fate.


Subject(s)
Biomimetic Materials/pharmacology , Collagen/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Integrins/metabolism , Peptides/chemical synthesis , Peptides/pharmacology , Amino Acid Sequence , Cell Adhesion/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Fibroblasts/metabolism , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Humans , Molecular Sequence Data , Peptides/chemistry , Protein Structure, Secondary , Signal Transduction/drug effects , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
13.
Biopolymers ; 90(6): 816-23, 2008.
Article in English | MEDLINE | ID: mdl-18846567

ABSTRACT

Development of artificial collagens to replace the animal-derived collagens presents a challenge in the formation of safer and functional biomaterials. We report here the development of collagen-like gels by means of the self-assembly of chemically synthesized peptides. The peptides are disulfide-linked trimers of collagenous Gly-X-Y triplet repeats with self-complementary shapes. Upon cooling the peptide solutions, hydrogels of peptide supramolecules are formed by spontaneous intermolecular triple helix formation. The thermal gel-sol transition appeared to be reversible, and the transition temperatures were found to be tunable by the design of the peptides. Our systems for the formation of artificial collagen-like gels will offer possibilities for novel types of biomaterials.


Subject(s)
Collagen/chemistry , Peptides/chemistry , Amino Acid Sequence , Chromatography, High Pressure Liquid , Collagen/chemical synthesis , Drug Stability , Gels , Hydrogel, Polyethylene Glycol Dimethacrylate , Molecular Sequence Data , Oligopeptides/chemistry , Thermodynamics
14.
J Biol Chem ; 281(16): 11177-85, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16484215

ABSTRACT

The endoplasmic reticulum-resident chaperone heat-shock protein 47 (HSP47) plays an essential role in procollagen biosynthesis. The function of HSP47 relies on its specific interaction with correctly folded triple-helical regions comprised of Gly-Xaa-Yaa repeats, and Arg residues at Yaa positions have been shown to be important for this interaction. The amino acid at the Yaa position (Yaa(-3)) in the N-terminal-adjoining triplet containing the critical Arg (defined as Arg(0)) was also suggested to be directly recognized by HSP47 (Koide, T., Asada, S., Takahara, Y., Nishikawa, Y., Nagata, K., and Kitagawa, K. (2006) J. Biol. Chem. 281, 3432-3438). Based on this finding, we examined the relationship between the structure of Yaa(-3) and HSP47 binding using synthetic collagenous peptides. The results obtained indicated that the structure of Yaa(-3) determined the binding affinity for HSP47. Maximal binding was observed when Yaa(-3) was Thr. Moreover, the required relative spatial arrangement of these key residues in the triple helix was analyzed by taking advantage of heterotrimeric collagen-model peptides, each of which contains one Thr(-3) and one Arg(0). The results revealed that HSP47 recognizes the Yaa(-3) and Arg(0) residues only when they are on the same peptide strand. Taken together, the data obtained led us to define the HSP47-binding structural epitope in the collagen triple helix and also define the HSP47-binding motif in the primary structure. A motif search against human protein database predicted candidate clients for this molecular chaperone. The search result indicated that not all collagen family proteins require the chaperoning by HSP47.


Subject(s)
Collagen/chemistry , HSP47 Heat-Shock Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Arginine/chemistry , Binding, Competitive , Cell Line , Circular Dichroism , Computational Biology , Databases, Protein , Endoplasmic Reticulum/metabolism , Epitopes/chemistry , Escherichia coli/metabolism , Glutathione Transferase/metabolism , HSP47 Heat-Shock Proteins/metabolism , Humans , Inhibitory Concentration 50 , Mice , Models, Molecular , Molecular Chaperones/chemistry , Molecular Sequence Data , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Software , Temperature , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...