Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 176(6): 1477-1489.e14, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30827683

RESUMEN

Receptor clustering on the cell membrane is critical in the signaling of many immunoreceptors, and this mechanism has previously been attributed to the extracellular and/or the intracellular interactions. Here, we report an unexpected finding that for death receptor 5 (DR5), a receptor in the tumor necrosis factor receptor superfamily, the transmembrane helix (TMH) alone in the receptor directly assembles a higher-order structure to drive signaling and that this structure is inhibited by the unliganded ectodomain. Nuclear magnetic resonance structure of the TMH in bicelles shows distinct trimerization and dimerization faces, allowing formation of dimer-trimer interaction networks. Single-TMH mutations that disrupt either trimerization or dimerization abolish ligand-induced receptor activation. Surprisingly, proteolytic removal of the DR5 ectodomain can fully activate downstream signaling in the absence of ligand. Our data suggest a receptor activation mechanism in which binding of ligand or antibodies to overcome the pre-ligand autoinhibition allows TMH clustering and thus signaling.


Asunto(s)
Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Apoptosis , Línea Celular Tumoral , Membrana Celular/metabolismo , Células HEK293 , Humanos , Ligandos , Modelos Moleculares , Mutagénesis Sitio-Dirigida/métodos , Unión Proteica , Proteolisis , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/química , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/ultraestructura , Transducción de Señal
2.
Angew Chem Int Ed Engl ; 62(27): e202302805, 2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-36961368

RESUMEN

One of the key challenges of improving clinical outcomes of antibody drug conjugates (ADCs) is overcoming cancer resistance to the antibody and/or drug components of ADCs, and hence the need for ADC platforms with high combinatory flexibility. Here, we introduce the use of self-assembled left-handed DNA (L-DNA) oligonucleotides to link combinatory single-domain antibodies and toxin payloads for tunable and adaptive delivery of ADCs. We demonstrate that the method allows convenient construction of a library of ADCs with multi-specific targeting, multi-specific payloads, and exact drug-antibody ratio. The newly constructed ADCs with L-DNA scaffold showed favorable properties of in vitro cell cytotoxicity and in vivo suppression and eradication of solid tumors. Collectively, our data suggest that the L-DNA based modular ADC (MADC) platform is a viable option for generating therapeutic ADCs and for potentially expanding ADC therapeutic window via multi-specificity.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Neoplasias , Humanos , Anticuerpos , ADN , Antineoplásicos/farmacología
3.
Nanomedicine ; 37: 102443, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34303839

RESUMEN

Neoantigen-based personalized vaccination has emerged as a viable method for tumor immunotherapy. Here we set up a DNA-based neoantigen vaccine platform with comprehensive identification of individual somatic mutations using whole-exome sequencing (WES) and RNA-seq, bioinformatic prediction of neo-epitopes, dendritic cell (DC)-based efficacy prevalidation of vaccine candidates, optimization of the DNA vaccine and its nanocarrier and adjuvant, and preparation of a liposome-encapsulated multiepitope DNA vaccine. The DNA vaccine was efficiently uptaken by DCs and induced effective immune response against mouse melanoma cells, leading to significant inhibition of melanoma tumor growth and reduction of lung metastasis in a mouse model. Numerous intratumoral infiltrated CD8+ T-cells with specific in vitro killing ability towards melanoma cells were identified. Our study offers evidence that a multiepitope neoantigen DNA vaccine in a nanocarrier can be exploited for personalized tumor immunotherapy and as a reliable prevalidation approach for rapid enrichment of effective neoantigens.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Inmunoterapia , Melanoma/terapia , Medicina de Precisión , Vacunas de ADN/farmacología , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Epítopos/genética , Epítopos/inmunología , Humanos , Liposomas/química , Liposomas/farmacología , Melanoma/inmunología , Melanoma/patología , Ratones , Mutación/genética , Nanopartículas/química , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Secuenciación del Exoma
4.
Bioconjug Chem ; 31(12): 2767-2778, 2020 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-33237767

RESUMEN

Despite the significant therapeutic advances in T-cell immunotherapy, many malignancies remain unresponsive, which might be because of the negative regulation of T cells by the tumor microenvironment (TME). T cells discriminate tumor cells and normal cells through T-cell receptors (TCRs); therefore, we generated a novel type of TCR-drug conjugates (TDCs) by referring antibody-drug conjugations (ADCs) to overcome the effects of the TME on T cells while preserving the specificity of TCR for tumor recognition. We selected HLA-A2/NY-ESO-1157-165 (peptide NY-ESO-1157-165 in complex with human leukocyte antigen serotype HLA-A*02:01) as the antigen and the antigen-specific TCR (1G4113) as the carrier. By sortase A-mediated ligation, we obtained three NY-TCR-vcMMAEs and further studied their properties, antitumor activity, and toxicity in vitro and in vivo. We found that all the NY-TCR-vcMMAEs had high endocytosis efficiency and specifically killed HLA-A2/NY-ESO-1157-165 positive tumor cells. In xenograft models, one of the TDCs, NY-TCR-2M, was effectively and specifically distributed into tumor tissues and inhibited tumor growth without inducing obvious toxicity. Our results demonstrated that TCRs can be carriers of toxic payloads and that the TDCs thus formed can specifically inhibit tumor growth, neglecting the immune microenvironment.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígenos de Superficie/inmunología , Proliferación Celular/efectos de los fármacos , Inmunoconjugados/farmacología , Espacio Intracelular/efectos de los fármacos , Proteínas de la Membrana/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Humanos , Inmunoconjugados/inmunología , Inmunoconjugados/metabolismo , Inmunoterapia , Espacio Intracelular/metabolismo , Ratones
5.
Small ; 13(6)2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27873460

RESUMEN

Antibody-drug conjugate (ADC) targeting antigens expressed on the surface of tumor cells are an effective approach for delivering drugs into the cells via antigen-mediated endocytosis. One of the well-known tumor antigens, the CD20 of B-lymphocyte, has long been suggested to be noninternalizing epitope, and is thus not considered a desirable target for ADCs. Here, sortase A (srtA)-mediated transpeptidation is used to specifically conjugate triple glycine-modified monomethyl auristatin E (MMAE), a highly toxic antimitotic agent, to anti-CD20 ofatumumab (OFA) equipped with a short C-terminal LPETG (5 amino acids) tag at heavy chain (HL), which generates ADCs that show extremely strong potency in killing CD20 positive cancer cells. One of the srtA-generated ADCs with a cleavable dipeptide linker (valine-citrulline, vc), OFA-HL-vcMMAE, shows IC50 values ranging from 5 pg mL-1 to 4.1 ng mL-1 against CD20+ lymphoma cells. Confocal laser scanning microscopy confirms that OFA-HL-vcMMAE internalization by Ramos cells is significantly improved compared to OFA alone, consistent with the high antitumor activity of the new ADC. OFA-HL-vcMMAE, at 5 mg kg-1 dose, is able to eliminate tumors with mean volume ≈400 mm3 while no obvious drug-related toxicity is observed. The results show that srtA-generated OFA-MMAE conjugate system provides a viable strategy for targeting CD20+ B lineage lymphomas.


Asunto(s)
Aminoaciltransferasas/metabolismo , Antígenos CD20/metabolismo , Proteínas Bacterianas/metabolismo , Linaje de la Célula , Cisteína Endopeptidasas/metabolismo , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/patología , Oligopéptidos/uso terapéutico , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Biocatálisis , Muerte Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Endocitosis/efectos de los fármacos , Humanos , Espectrometría de Masas , Ratones , Oligopéptidos/síntesis química , Oligopéptidos/química , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Bioconjug Chem ; 28(8): 2180-2189, 2017 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-28697305

RESUMEN

Although TRAIL (tumor-necrosis-factor (TNF)-related apoptosis-inducing ligand) has been considered a promising broad-spectrum antitumor agent, its further application was limited by poor drug delivery and TRAIL-resistant tumors. A three-step drug delivery strategy was applied to TRAIL for solving these two obstacles in the form of PEG-TRAIL-MMAE (Monomethyl Auristatin E). PEGylation of TRAIL in the first step was carried out to improve its in vivo pharmacokinetics, while the interaction between TRAIL conjugates with death receptors in the second step was designed to activate the TRAIL extrinsic apoptosis pathway, and the further release of MMAE from the lysosome was the third step for introducing another apoptosis pathway to overcome TRAIL resistance in some tumors. Herein, in order to reach a balance among the three steps, the PEG/MMAE ratio was optimized for PEG-TRAIL-MMAE conjugates. PEG-TRAIL-MMAE conjugates with various PEG/MMAE ratios were prepared and compared with each other regarding their pharmacokinetics (PK) and pharmacodynamics (PD). As a result, PEG-TRAIL-MMAE conjugates with a PEG/MMAE ratio of 1:2 showed prolonged half-life in rats (6.8 h), and the best antitumor activity in vitro (IC50 0.31 nM) and in vivo while no sign of toxicity in xenograft models, suggesting it as a promising multistep drug delivery and antitumor strategy after optimization.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Portadores de Fármacos/química , Oligopéptidos/química , Polietilenglicoles/química , Ligando Inductor de Apoptosis Relacionado con TNF/química , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Animales , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Transformación Celular Neoplásica , Femenino , Ligandos , Masculino , Ratones , Ratas , Ligando Inductor de Apoptosis Relacionado con TNF/farmacocinética , Distribución Tisular
7.
Biotechnol Appl Biochem ; 64(5): 627-637, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27572239

RESUMEN

Human serum albumin (HSA) and interleukin-1 receptor antagonist (IL1Ra) fusion protein is a potential long-acting drug in the treatment of type 2 diabetes. Previously, the expression level of HSA/IL1Ra in Pichia pastoris was successfully improved by increasing the gene copy number and coexpression with chaperone (protein disulfide isomerase) in our laboratory. However, the overexpression strain resulted in low production of high- cell-density fermentation. In this study, the culture medium was optimized in both flask and fermenter, and the optimum culture medium notably increased the productivity and stability of HSA/IL1Ra. To further improve the expression, response surface methodology was used to further optimize the culture condition through modeling three selected parameters (induction pH, induction temperature [T], and maximum methanol feed rate [Vm ]). The maximum yield of HSA/IL1Ra reached 1.1 g/L (10-fold higher than original fermentation condition) under the optimized culture condition (pH 7.0, T = 29 â„ƒ and Vm = 4.82 mL/L/H) in a 5-L fermenter. In addition, the degradation position of HSA/IL1Ra during fermentation was determined to be K571, serving as a potential target for genetic modification strategies to reduce the degradation. Finally, the in vivo activity study showed that HSA/IL1Ra maintained the therapeutic effect of IL1Ra in type 2 diabetes model rats meanwhile reducing the frequency of administration.


Asunto(s)
Hipoglucemiantes , Proteína Antagonista del Receptor de Interleucina 1 , Pichia/metabolismo , Proteínas Recombinantes de Fusión , Albúmina Sérica , Animales , Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2 , Humanos , Hiperglucemia , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacocinética , Hipoglucemiantes/farmacología , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/farmacocinética , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Masculino , Ratones , Ratones Endogámicos ICR , Pichia/genética , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/farmacología , Proyectos de Investigación , Albúmina Sérica/metabolismo , Albúmina Sérica/farmacocinética , Albúmina Sérica/farmacología
8.
Methods Mol Biol ; 2793: 41-54, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38526722

RESUMEN

Resistance to therapeutic antibodies caused by on-target point mutations is a major obstacle in anticancer therapy, creating an "unmet clinical need." To tackle this problem, researchers are developing new generations of antibody drugs that can overcome the resistance mechanisms of existing agents. We have previously reported a structure-guided and phage-assisted evolution (SGAPAE) approach to evolve cetuximab, a therapeutic antibody, to effectively reverse the resistance driven by EGFRS492R or EGFRG465R mutations, without changing the binding epitope or compromising the antibody efficacy. In this protocol, we provide detailed instructions on how to use the SGAPAE approach to evolve cetuximab, which can also be applied to other therapeutic antibodies for reversing on-target point mutation-mediated resistance. The protocol consists of four steps: structure preparation, computational prediction, phage display library construction, and antibody candidate selection.


Asunto(s)
Anticuerpos Monoclonales , Bacteriófagos , Cetuximab , Mutación Puntual , Receptores ErbB/metabolismo , Bacteriófagos/metabolismo , Anticuerpos Monoclonales Humanizados/genética
9.
Biomedicines ; 12(4)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38672132

RESUMEN

Antibody-based bispecific T cell engagers (TCEs) that redirect T cells to kill tumor cells have shown a promising therapeutic effect on hematologic malignancies. However, tumor-specific targeting is still a challenge for TCEs, impeding the development of TCEs for solid tumor therapy. The major histocompatibility complex (MHC) presents almost all intracellular peptides (including tumor-specific peptides) on the cell surface to be scanned by the TCR on T cells. With the premise of choosing optimal peptides, the final complex peptide-MHC could be the tumor-specific target for TCEs. Here, a novel TCR-directed format of a TCE targeting peptide-MHC was designed named IgG-T-TCE, which was modified from the IgG backbone and prepared in a mammalian cell expression system. The recombinant IgG-T-TCE-NY targeting NY-ESO-1157-165/HLA-A*02:01 could be generated in HEK293 cells with a glycosylated TCR and showed potency in T cell activation and redirecting T cells to specifically kill target tumor cells. We also found that the in vitro activity of IgG-T-TCE-NY could be leveraged by various anti-CD3 antibodies and Fc silencing. The IgG-T-TCE-NY efficiently inhibited tumor growth in a tumor-PBMC co-engrafted mouse model without any obvious toxicities.

10.
Signal Transduct Target Ther ; 7(1): 39, 2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-35132063

RESUMEN

Monoclonal antibodies constitute a promising class of targeted anticancer agents that enhance natural immune system functions to suppress cancer cell activity and eliminate cancer cells. The successful application of IgG monoclonal antibodies has inspired the development of various types of therapeutic antibodies, such as antibody fragments, bispecific antibodies, and antibody derivatives (e.g., antibody-drug conjugates and immunocytokines). The miniaturization and multifunctionalization of antibodies are flexible and viable strategies for diagnosing or treating malignant tumors in a complex tumor environment. In this review, we summarize antibodies of various molecular types, antibody applications in cancer therapy, and details of clinical study advances. We also discuss the rationale and mechanism of action of various antibody formats, including antibody-drug conjugates, antibody-oligonucleotide conjugates, bispecific/multispecific antibodies, immunocytokines, antibody fragments, and scaffold proteins. With advances in modern biotechnology, well-designed novel antibodies are finally paving the way for successful treatments of various cancers, including precise tumor immunotherapy, in the clinic.


Asunto(s)
Anticuerpos Biespecíficos , Antineoplásicos Inmunológicos , Inmunoconjugados , Neoplasias , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/uso terapéutico , Humanos , Inmunoconjugados/inmunología , Inmunoconjugados/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología
11.
Nat Commun ; 13(1): 4431, 2022 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-35907884

RESUMEN

Acquired resistance to cetuximab in colorectal cancers is partially mediated by the acquisition of mutations located in the cetuximab epitope in the epidermal growth factor receptor (EGFR) ectodomain and hinders the clinical application of cetuximab. We develop a structure-guided and phage-assisted evolution approach for cetuximab evolution to reverse EGFRS492R- or EGFRG465R-driven resistance without altering the binding epitope or undermining antibody efficacy. Two evolved cetuximab variants, Ctx-VY and Ctx-Y104D, exhibit a restored binding ability with EGFRS492R, which harbors the most common resistance substitution, S492R. Ctx-W52D exhibits restored binding with EGFR harboring another common cetuximab resistance substitution, G465R (EGFRG465R). All the evolved cetuximab variants effectively inhibit EGFR activation and downstream signaling and induce the internalization and degradation of EGFRS492R and EGFRG465R as well as EGFRWT. The evolved cetuximab variants (Ctx-VY, Ctx-Y104D and Ctx-W52D) with one or two amino acid substitutions in the complementarity-determining region inherit the optimized physical and chemical properties of cetuximab to a great extent, thus ensuring their druggability. Our data collectively show that structure-guided and phage-assisted evolution is an efficient and general approach for reversing receptor mutation-mediated resistance to therapeutic antibody drugs.


Asunto(s)
Antineoplásicos , Bacteriófagos , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/farmacología , Bacteriófagos/genética , Línea Celular Tumoral , Cetuximab/farmacología , Cetuximab/uso terapéutico , Resistencia a Antineoplásicos/genética , Epítopos
12.
Cancers (Basel) ; 13(18)2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34572769

RESUMEN

Bispecific antibodies (BsAbs) for T cell engagement have shown great promise in cancer immunotherapy, and their clinical applications have been proven in treating hematological malignance. Bispecific antibody binding fragment (BiFab) represents a promising platform for generating non-Fc bispecific antibodies. However, the generation of BiFab is still challenging, especially by means of chemical conjugation. More conjugation strategies, e.g., enzymatic conjugation and modular BiFab preparation, are needed to improve the robustness and flexibility of BiFab preparation. We successfully used chemo-enzymatic conjugation approach to generate bispecific antibody (i.e., BiFab) with Fabs from full-length antibodies. Paired click handles (e.g., N3 and DBCO) was introduced to the C-terminal LPETG tag of Fabs via sortase A mediated transpeptidation, followed by site-specific conjugation between two click handle-modified Fabs for BiFab generation. Both BiFabCD20/CD3 (EC50 = 0.26 ng/mL) and BiFabHer2/CD3 exhibited superior efficacy in mediating T cells, from either PBMC or ATC, to kill target tumor cell lines while spared antigen-negative tumor cells in vitro. The BiFabCD20/CD3 also efficiently inhibited CD20-positive tumor growth in mouse xenograft model. We have established a facile sortase A-mediated click handle installation to generate homogeneous and functional BiFabs. The exemplary BiFabs against different targets showed superior efficacy in redirecting and activating T cells to specifically kill target tumor cells, demonstrating the robustness of sortase A-mediated "bio-click" chemistry in generating various potent BiFabs. This approach also holds promise for further efficient construction of a Fab derivative library for personalized tumor immunotherapy in the future.

13.
Biomedicines ; 9(7)2021 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-34356854

RESUMEN

The specific recognition of T cell receptors (TCR) and peptides presented by human leukocyte antigens (pHLAs) is the core step for T cell triggering to execute anti-tumor activity. However, TCR assembly and soluble expression are challenging, which precludes the broad use of TCR in tumor therapy. Herein, we used heterodimeric Fc to assist in the correct assembly of TCRs to achieve the stable and soluble expression of several TCRs in mammalian cells, and the soluble TCRs enable us to yield novel bispecific T cell engagers (TCR/aCD3) through pairing them with an anti-CD3 antibody. The NY-ESO-1/LAGE-1 targeted TCR/aCD3 (NY-TCR/aCD3) that we generated can redirect naïve T cells to specific lysis antigen-positive tumor cells, but the potency of the NY-TCR/aCD3 was disappointing. Furthermore, we found that the activation of T cells by NY-TCR/aCD3 was mild and unabiding, and the activity of NY-TCR/aCD3 could be significantly improved when we replaced naïve T cells with pre-activated T cells. Therefore, we employed the robust T cell activation ability of staphylococcal enterotoxin C2 (SEC2) to optimize the activity of NY-TCR/aCD3. Moreover, we found that the secretions of SEC2-activated T cells can promote HLA-I expression and thus increase target levels, which may further contribute to improving the activity of NY-TCR/aCD3. Our study described novel strategies for soluble TCR expression, and the optimization of the generation and potency of TCR/aCD3 provided a representative for us to fully exploit TCRs for the precision targeting of cancers.

14.
Front Cell Dev Biol ; 8: 569684, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33163490

RESUMEN

Receptors in the tumor necrosis factor receptor superfamily (TNFRSF) regulate proliferation of immune cells or induce programmed cell death, and many of them are candidates for antibody-based immunotherapy. Previous studies on several death receptors in the TNFRSF including Fas, p75NTR, and DR5 showed that the transmembrane helix (TMH) of these receptors can specifically oligomerize and their oligomeric states have direct consequences on receptor activation, suggesting a much more active role of TMH in receptor signaling than previously appreciated. Here, we report the structure of the TMH of TNFR1, another well studied member of the TNFRSF, in neutral bicelles that mimic a lipid bilayer. We find that TNFR1 TMH forms a defined trimeric complex in bicelles, and no evidences of higher-order clustering of trimers have been detected. Unexpectedly, a conserved proline, which is critical for Fas TMH trimerization, does not appear to play an important role in TNFR1 TMH trimerization, which is instead mediated by a glycine near the middle of the TMH. Further, TNFR1 TMH trimer shows a larger hydrophobic core than that of Fas or DR5, with four layers of hydrophobic interaction along the threefold axis. Comparison of the TNFR1 TMH structure with that of Fas and DR5 reveals reassuring similarities that have functional implications but also significant structural diversity that warrants systematic investigation of TMH oligomerization property for other members of the TNFRSF.

15.
Adv Sci (Weinh) ; 7(2): 1900973, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31993277

RESUMEN

Targeting T-cells against cancer cells is a direct means of treating cancer, and has already shown great responses in clinical treatment of B-cell malignancies. A simple way to redirect T-cells to cancer cells is by using multispecific antibody (MsAb) that contains different arms for specifically "grabbing" the T-cells and cancer cells; as such, the T-cells are activated upon target engagement and the killing begins. Here, a nucleic acid mediated protein-protein assembly (NAPPA) approach is implemented to construct a MsAb for T-cell engaging and tumor killing. Anti -CD19 and -CD3 single-chain variable fragments (scFvs) are conjugated to different l-DNAs with sequences that form the Holliday junction, thus allowing spontaneous assembly of homogeneous protein-DNA oligomers containing two anti-CD19 and one anti-CD3 scFvs. The new MsAb shows strong efficacy in inducing Raji tumor cell cytotoxicity in the presence of T-cells with EC50 ≈ 0.2 × 10-9 m; it also suppresses tumor growth in a Raji xenograft mouse model. The data indicates that MsAbs assembled from protein-DNA conjugates are effective macromolecules for directing T-cells for tumor killing. The modular nature of the NAPPA platform allows rapid generation of complex MsAbs from simple antibody fragments, while offering a general solution for preparing antibodies with high-order specificity.

16.
Asian J Pharm Sci ; 15(6): 777-785, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33363632

RESUMEN

Limited clinical application of antibody-drug conjugates (ADCs) targeting tumor associated antigens (TAAs) is usually caused by on-target off-tumor side effect. Tumor-specific mutant antigens (TSMAs) only expressed in tumor cells which are ideal targets for ADCs. In addition, intracellular somatic mutant proteins can be presented on the cell surface by human leukocyte antigen class I (HLA I)molecules forming tumor-specific peptide/HLA I complexes. KRAS G12V mutation frequently occurred in varied cancer and was verified as a promising target for cancer therapy. In this study, we generated two TCR-mimic antibody-drug conjugates (TCRm-ADCs), 2E8-MMAE and 2A5-MMAE, targeting KRAS G12V/HLA-A*0201 complex, which mediated specific antitumor activity in vitro and in vivo without obvious toxicity. Our findings are the first time validate the strategy of TCRm-ADCs targeting intracellular TSMAs, which improves the safety of antibody-based drugs and provides novel strategy for precision medicine in cancer therapy.

17.
Eur J Pharm Sci ; 134: 81-92, 2019 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-30986472

RESUMEN

Antibody fragments, as the products of engineered antibodies, exhibit great potential for cancer therapy and imaging. Antibody fragment drug conjugates (AFDCs), which conjugate the highly specific, low-immunity and small-sized antibody fragments with cytotoxic payloads, can overcome the limitations of traditional IgG format drugs in cancer therapy. In this study, a commercialized anti-CD20 monoclonal antibody, ofatumumab (OFA), was applied to generate two site-specific monomethyl auristain E (MMAE)-conjugated AFDCs (Fab-vcMMAE, Fab-CH3mut-vcMMAE) by Sortase A mediated transpeptidation. Compared with OFA-vcMMAE, the two AFDCs maintained most of the binding affinity and the ability of internalization. In vitro studies revealed that Fab-vcMMAE and OFA-vcMMAE had almost identical IC50 values against CD20-positive cell lines, while Fab-CH3-vcMMAE had a lower anti-tumor activity. In vivo studies showed that Fab-vcMMAE had a significantly higher maximum tolerated dose (MTDs), a 30-fold shorter half-life, and slightly lower antitumor activity within the MTDs than OFA-vcMMAE. The distribution study showed that both of the Fab and Fab-CH3mut had higher penetration rates into the tumors than OFA in a xenograft model. Additionally, no obvious difference in tumor drug accumulation was found between the Fab and OFA groups after the penetration process, but the Fab-CH3mut group exhibited less tumor drug accumulation, possibly contributing to the inferior anti-tumor activity of Fab-CH3mut-vcMMAE in vivo. Overall, we preliminarily demonstrated the characteristics of AFDCs by studying OFA-based AFDCs. Our results revealed that Fab is a promising carrier of MMAE to enhance the anti-tumor activity and increase the safety profile compared with OFA.


Asunto(s)
Antineoplásicos/farmacología , Inmunoconjugados/farmacología , Fragmentos de Inmunoglobulinas/farmacología , Animales , Anticuerpos Monoclonales Humanizados/farmacocinética , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos CD20 , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Inmunoconjugados/farmacocinética , Ratones , Ratones Endogámicos BALB C , Oligopéptidos , Rituximab
18.
Onco Targets Ther ; 12: 11597-11608, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31920346

RESUMEN

BACKGROUND: The aberrant expression of HER2 is highly associated with tumour occurrence and metastasis, therefore HER2 is extensively targeted for tumour immunotherapy. For example, trastuzumab and pertuzumab are FDA-approved monoclonal antibodies that target HER2-positive tumour cells. Despite their advances in clinical applications, emerging resistance to these two HER2-targeting antibodies has hindered their further application. Somatic mutations in HER2 receptor have been identified as one of the major reasons for resistance to anti-HER2 antibodies. METHODS: We analysed the frequency of somatic mutations in various tumour types based on TCGA and COSMIC databases. Then, the effect of the most frequent mutation (S310F) on the interaction between pertuzumab and HER2 was analysed by molecular modelling analysis. The effect of the S310F mutation was further evaluated through multiple in vitro binding experiments and antitumour activity assays. RESULTS: We found through bioinformatics analysis that S310F, an activating mutation in the HER2 extracellular domain, was the most frequent mutation in HER2. The S310F mutation was shown to confer resistance of HER2-positive tumour cells to pertuzumab treatment. With molecular modelling analysis, we confirmed the possibility that the S310F mutation might disrupt the interaction between pertuzumab and HER2 as a result of a significant change in the critical residue S310. Further functional analyses revealed that the S310F mutation completely abolished pertuzumab binding to HER2 receptor and inhibited pertuzumab antitumour efficacy. CONCLUSION: We demonstrated the loss-of-function mechanism underlying pertuzumab resistance in HER2-positive tumour cells bearing the S310F mutation.

19.
Structure ; 26(4): 627-634.e4, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29551287

RESUMEN

The p7 protein of the hepatitis C virus (HCV) can oligomerize in membrane to form cation channels. Previous studies showed that the channel assembly in detergent micelles adopts a unique flower-shaped oligomer, but the unusual architecture also presented problems for understanding how this viroporin resides in the membrane. Moreover, the oligomeric state of p7 remains controversial, as both hexamer and heptamer have been proposed. Here we address the above issues using p7 reconstituted in bicelles that mimic a lipid bilayer. We found, using a recently developed oligomer-labeling method, that p7 forms hexamers in the bicelles. Solvent paramagnetic relaxation enhancement analyses showed that the bilayer thickness around the HCV ion channel is substantially smaller than expected, and thus a significant portion of the previously assigned membrane-embedded region is solvent exposed. Our study provides an effective approach for characterizing the transmembrane partition of small ion channels in near lipid bilayer environment.


Asunto(s)
Materiales Biomiméticos/química , Dimiristoilfosfatidilcolina/química , Hepacivirus/química , Canales Iónicos/química , Membrana Dobles de Lípidos/química , Éteres Fosfolípidos/química , Proteínas Virales/química , Secuencias de Aminoácidos , Sitios de Unión , Materiales Biomiméticos/metabolismo , Clonación Molecular , Cristalografía por Rayos X , Dimiristoilfosfatidilcolina/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Hepacivirus/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Membrana Dobles de Lípidos/metabolismo , Modelos Moleculares , Éteres Fosfolípidos/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
20.
Biomaterials ; 178: 158-169, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29933102

RESUMEN

Most tumor-associated proteins are located inside tumor cells and thus are not accessible to current marketed therapeutic monoclonal antibodies or their cytotoxic conjugates. Human leukocyte antigen (HLA) class I can present peptides derived from intracellular tumor-associated proteins and somatically mutated proteins on the cell's surface, forming an HLA/peptide complex as tumor-specific antigens for T cell receptor (TCR) recognition. Therefore, HLA-mediated presentation of intracellular tumor antigen peptides provides a viable way to distinguish tumor cells from normal cells, which is important for broadening antigen selection, especially for antibody-drug conjugates (ADCs) regarding their highly cytotoxic payload. We applied sortase A-mediated conjugation to develop TCR-like ADCs (i.e., EA1 HL-vcMMAE) targeting intracellular MART-1 protein, a melanocyte-differentiating antigen specific for metastatic melanomas, via the cell surface HLA-A2/MART-126-35 peptide complex. Homogenous EA1 HL-vcMMAE (drug to antibody ratio of 4) efficiently eliminated melanoma cells in xenograft mouse models with no obvious toxicity at the therapeutic dosage. Trametinib, an MEK inhibitor serving as an HLA expression enhancing agent, augmented the TL-ADCs' efficacy both in vitro and in vivo by upregulating MART-126-35 peptide presentation, thus providing a strategy for overcoming the limitation of antigen presentation level for TL-ADCs. Hence, our findings validate the strategy of using sortase A-generated TL-ADCs to target tumor-specific intracellular proteins, with or without agents present, to increase presenting TCR epitope peptides.


Asunto(s)
Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Inmunoconjugados/metabolismo , Espacio Intracelular/metabolismo , Antígeno MART-1/metabolismo , Melanoma/patología , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Especificidad de Anticuerpos/efectos de los fármacos , Presentación de Antígeno/efectos de los fármacos , Antineoplásicos/farmacología , Línea Celular Tumoral , Endocitosis/efectos de los fármacos , Femenino , Antígenos de Histocompatibilidad Clase I/metabolismo , Ratones SCID , Péptidos/química , Unión Proteica/efectos de los fármacos , Piridonas/farmacología , Pirimidinonas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA