Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(32): e2204078119, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35914154

RESUMEN

Peptide-based cancer vaccines are widely investigated in the clinic but exhibit modest immunogenicity. One approach that has been explored to enhance peptide vaccine potency is covalent conjugation of antigens with cell-penetrating peptides (CPPs), linear cationic and amphiphilic peptide sequences designed to promote intracellular delivery of associated cargos. Antigen-CPPs have been reported to exhibit enhanced immunogenicity compared to free peptides, but their mechanisms of action in vivo are poorly understood. We tested eight previously described CPPs conjugated to antigens from multiple syngeneic murine tumor models and found that linkage to CPPs enhanced peptide vaccine potency in vivo by as much as 25-fold. Linkage of antigens to CPPs did not impact dendritic cell activation but did promote uptake of linked antigens by dendritic cells both in vitro and in vivo. However, T cell priming in vivo required Batf3-dependent dendritic cells, suggesting that antigens delivered by CPP peptides were predominantly presented via the process of cross-presentation and not through CPP-mediated cytosolic delivery of peptide to the classical MHC class I antigen processing pathway. Unexpectedly, we observed that many CPPs significantly enhanced antigen accumulation in draining lymph nodes. This effect was associated with the ability of CPPs to bind to lymph-trafficking lipoproteins and protection of CPP-antigens from proteolytic degradation in serum. These two effects resulted in prolonged presentation of CPP-peptides in draining lymph nodes, leading to robust T cell priming and expansion. Thus, CPPs can act through multiple unappreciated mechanisms to enhance T cell priming that can be exploited for cancer vaccines with enhanced potency.


Asunto(s)
Vacunas contra el Cáncer , Péptidos de Penetración Celular , Inmunogenicidad Vacunal , Ganglios Linfáticos , Animales , Presentación de Antígeno , Antígenos , Vacunas contra el Cáncer/inmunología , Péptidos de Penetración Celular/farmacología , Reactividad Cruzada , Células Dendríticas/inmunología , Ganglios Linfáticos/inmunología , Ratones , Linfocitos T/inmunología , Vacunas de Subunidad/inmunología
2.
Biomacromolecules ; 22(7): 2850-2863, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34156837

RESUMEN

Over the past decade, extensive optimization of polymeric cell-penetrating peptide (CPP) mimics (CPPMs) by our group has generated a substantial library of broadly effective carriers which circumvent the need for covalent conjugation often required by CPPs. In this study, design rules learned from CPPM development were applied to reverse-engineer the first library of simple amphiphilic block copolypeptides for non-covalent protein delivery, namely, poly(alanine-block-arginine), poly(phenylalanine-block-arginine), and poly(tryptophan-block-arginine). This new CPP library was screened for enhanced green fluorescent protein and Cre recombinase delivery alongside a library of CPPMs featuring equivalent side-chain configurations. Due to the added hydrophobicity imparted by the polymer backbone as compared to the polypeptide backbone, side-chain functionality was not a universal predictor of carrier performance. Rather, overall carrier hydrophobicity predicted the top performers for both internalization and activity of protein cargoes, regardless of backbone identity. Furthermore, comparison of protein uptake and function revealed carriers which facilitated high gene recombination despite remarkably low Cre internalization, leading us to formalize the concept of intracellular availability (IA) of the delivered cargo. IA, a measure of cargo activity per quantity of cargo internalized, provides valuable insight into the physical relationship between cellular internalization and bioavailability, which can be affected by bottlenecks such as endosomal escape and cargo release. Importantly, carriers with maximal IA existed within a narrow hydrophobicity window, more hydrophilic than those exhibiting maximal cargo uptake. Hydrophobicity may be used as a scaffold-independent predictor of protein uptake, function, and IA, enabling identification of new, effective carriers which would be overlooked by uptake-based screening methods.


Asunto(s)
Péptidos de Penetración Celular , Transporte Biológico , Péptidos de Penetración Celular/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Polímeros , Transporte de Proteínas
3.
Mol Pharm ; 16(6): 2462-2469, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31095395

RESUMEN

Delivering peptides and proteins with intracellular function represents a promising avenue for therapeutics, but remains a challenge due to the selective permeability of the plasma membrane. The successful delivery of cytosolically active proteins would enable many opportunities, including improved vaccine development through major histocompatibility complex (MHC) class I antigen display. Extended research using cell-penetrating peptides (CPPs) has aimed to facilitate intracellular delivery of exogenous proteins with some success. A new class of polymer-based mimics termed protein transduction domain mimics (PTDMs), which maintain the positive charge and amphiphilic nature displayed by many CPPs, was developed using a poly-norbornene-based backbone. Herein, we use a previously characterized PTDM to investigate delivery of the model antigen SIINFEKL into leukocytes. Peptide delivery into over 90% of CD14+ monocytes was detected in less than 15 min with nominal inflammatory cytokine response and high cell viability. The co-delivery of a TLR9 agonist and antigen using the PTDM into antigen-presenting cells in vitro showed presentation of SIINFEKL in association with MHC class I molecules, in addition to upregulation of classical differentiation markers revealing the ability of the PTDM to successfully deliver cargo intracellularly and show application in the field of immunotherapy.


Asunto(s)
Monocitos/metabolismo , Presentación de Antígeno/fisiología , Supervivencia Celular/fisiología , Péptidos de Penetración Celular/metabolismo , Células Dendríticas/metabolismo , Citometría de Flujo , Voluntarios Sanos , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Receptores de Lipopolisacáridos/metabolismo , Células THP-1 , Receptor Toll-Like 9/metabolismo
4.
Biochim Biophys Acta ; 1858(7 Pt A): 1443-50, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27039278

RESUMEN

Elucidating the predominant cellular entry mechanism for protein transduction domains (PTDs) and their synthetic mimics (PTDMs) is a complicated problem that continues to be a significant source of debate in the literature. The PTDMs reported here provide a well-controlled platform to vary molecular composition for structure activity relationship studies to further our understanding of PTDs, their non-covalent association with cargo, and their cellular internalization pathways. Specifically, several guanidine rich homopolymers, along with an amphiphilic block copolymer were used to investigate the relationship between structure and internalization activity in HeLa cells, both alone and non-covalently complexed with EGFP by flow cytometery and confocal imaging. The findings indicate that while changing the amount of positive charge on our PTDMs does not seem to affect the endosomal uptake, the presence of hydrophobicity appears to be a critical factor for the polymers to enter cells either alone, or with associated cargo.


Asunto(s)
Endocitosis/fisiología , Endosomas/metabolismo , Guanidinas/metabolismo , Peptidomiméticos/metabolismo , Polímeros/metabolismo , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/metabolismo , Citometría de Flujo , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Guanidinas/síntesis química , Células HeLa , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Peptidomiméticos/síntesis química , Polímeros/síntesis química , Estructura Terciaria de Proteína , Transporte de Proteínas , Electricidad Estática , Relación Estructura-Actividad
5.
Chemistry ; 23(28): 6858-6863, 2017 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-28370636

RESUMEN

Cell-penetrating peptides are an important class of molecules with promising applications in bioactive cargo delivery. A diverse series of guanidinium-containing polymeric cell-penetrating peptide mimics (CPPMs) with varying backbone chemistries was synthesized and assessed for delivery of both GFP and fluorescently tagged siRNA. Specifically, we examined CPPMs based on norbornene, methacrylate, and styrene backbones to determine how backbone structure impacted internalization of these two cargoes. Either charge content or degree of polymerization was held constant at 20, with diguanidinium norbornene molecules being polymerized to both 10 and 20 repeat units. Generally, homopolymer CPPMs delivered low amounts of siRNA into Jurkat T cells, with no apparent backbone dependence; however, by adding a short hydrophobic methyl methacrylate block to the guanidinium-rich methacrylate polymer, siRNA delivery to nearly the entire cell population was achieved. Protein internalization yielded similar results for most of the CPPMs, though the block polymer was unable to deliver proteins. In contrast, the styrene-based CPPM yielded the highest internalization for GFP (≈40 % of cells affected), showing that indeed backbone chemistry impacts protein delivery, specifically through the incorporation of an aromatic group. These results demonstrate that an understanding of how polymer structure affects cargo-dependent internalization is critical to designing new, more effective CPPMs.


Asunto(s)
Materiales Biocompatibles/síntesis química , Guanidina/química , Polímeros/química , Materiales Biocompatibles/química , Péptidos de Penetración Celular/química , Citometría de Flujo , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Células Jurkat , Metilmetacrilato/química , Norbornanos/química , ARN Interferente Pequeño/química , ARN Interferente Pequeño/metabolismo , Transfección/métodos
6.
Biomacromolecules ; 18(3): 819-825, 2017 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-28165726

RESUMEN

The use of proteins as biological tools and therapeutic agents is limited due to the fact that proteins do not effectively cross the plasma membrane of cells. Here, we report a novel class of protein transporter molecules based on protein transduction domain mimics (PTDMs) synthesized via ring opening metathesis polymerization (ROMP). The PTDMs reported here were specifically inspired by amphiphilic peptides known to deliver functional proteins into cells via noncovalent interactions between the peptide and the cargo. This contrasts with peptides like TAT, penetratin, and R9, which often require covalent fusion to their cargoes. Using the easily tunable synthetic ROMP platform, the importance of a longer hydrophobic segment with cationic guanidinium groups was established through the delivery of EGFP into Jurkat T cells. The most efficient of these protein transporters was used to deliver functional Cre Recombinase with ∼80% knockdown efficiency into hard to transfect human T cells. Additionally, a C-terminally deleted form of the transcription factor Runx1 (Runx1.d190) was delivered into primary murine splenocytes, producing a 2-fold increase in c-Myc mRNA production, showcasing the versatility of this platform to deliver biologically active proteins into hard to transfect cell types.


Asunto(s)
Biomimética , Transporte de Proteínas , Proteínas/química , Transfección , Animales , Sistemas de Liberación de Medicamentos , Proteínas Fluorescentes Verdes/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Integrasas/química , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Polimerizacion , Bazo/citología , Bazo/efectos de los fármacos , Linfocitos T/efectos de los fármacos
7.
Elife ; 122023 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-37548358

RESUMEN

Cancer immunotherapies, in particular checkpoint blockade immunotherapy (CBT), can induce control of cancer growth, with a fraction of patients experiencing durable responses. However, the majority of patients currently do not respond to CBT and the molecular determinants of resistance have not been fully elucidated. Mounting clinical evidence suggests that the clonal status of neoantigens (NeoAg) impacts the anti-tumor T cell response. High intratumor heterogeneity (ITH), where the majority of NeoAgs are expressed subclonally, is correlated with poor clinical response to CBT and poor infiltration with tumor-reactive T cells. However, the mechanism by which ITH blunts tumor-reactive T cells is unclear. We developed a transplantable murine lung cancer model to characterize the immune response against a defined set of NeoAgs expressed either clonally or subclonally to model low or high ITH, respectively. Here we show that clonal expression of a weakly immunogenic NeoAg with a relatively strong NeoAg increased the immunogenicity of tumors with low but not high ITH. Mechanistically we determined that clonal NeoAg expression allowed cross-presenting dendritic cells to acquire and present both NeoAgs. Dual NeoAg presentation by dendritic cells was associated with a more mature DC phenotype and a higher stimulatory capacity. These data suggest that clonal NeoAg expression can induce more potent anti-tumor responses due to more stimulatory dendritic cell:T cell interactions. Therapeutic vaccination targeting subclonally expressed NeoAgs could be used to boost anti-tumor T cell responses.


Asunto(s)
Reactividad Cruzada , Neoplasias Pulmonares , Animales , Ratones , Antígenos de Neoplasias/genética , Neoplasias Pulmonares/genética , Linfocitos T , Células Dendríticas
8.
ACS Cent Sci ; 9(9): 1835-1845, 2023 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-37780364

RESUMEN

Molecular vaccines comprising antigen peptides and inflammatory cues make up a class of therapeutics that promote immunity against cancer and pathogenic diseases but often exhibit limited efficacy. Here, we engineered an antigen peptide delivery system to enhance vaccine efficacy by targeting dendritic cells and mediating cytosolic delivery. The delivery system consists of the nontoxic anthrax protein, protective antigen (PA), and a single-chain variable fragment (scFv) that recognizes the XCR1 receptor on dendritic cells (DCs). Combining these proteins enabled selective delivery of the N-terminus of lethal factor (LFN) into XCR1-positive cross-presenting DCs. Incorporating immunogenic epitope sequences into LFN showed selective protein translocation in vitro and enhanced the priming of antigen-specific T cells in vivo. Administering DC-targeted constructs with tumor antigens (Trp1/gp100) into mice bearing aggressive B16-F10 melanomas improved mouse outcomes when compared to free antigen, including suppressed tumor growth up to 58% at 16 days post tumor induction (P < 0.0001) and increased survival (P = 0.03). These studies demonstrate that harnessing DC-targeting anthrax proteins for cytosolic antigen delivery significantly enhances the immunogenicity and antitumor efficacy of cancer vaccines.

9.
Nat Biomed Eng ; 7(9): 1113-1128, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37291434

RESUMEN

The effectiveness of chimaeric antigen receptor (CAR) T cell therapies for solid tumours is hindered by difficulties in the selection of an effective target antigen, owing to the heterogeneous expression of tumour antigens and to target antigen expression in healthy tissues. Here we show that T cells with a CAR specific for fluorescein isothiocyanate (FITC) can be directed against solid tumours via the intratumoural administration of a FITC-conjugated lipid-poly(ethylene)-glycol amphiphile that inserts itself into cell membranes. In syngeneic and human tumour xenografts in mice, 'amphiphile tagging' of tumour cells drove tumour regression via the proliferation and accumulation of FITC-specific CAR T cells in the tumours. In syngeneic tumours, the therapy induced the infiltration of host T cells, elicited endogenous tumour-specific T cell priming and led to activity against distal untreated tumours and to protection against tumour rechallenge. Membrane-inserting ligands for specific CARs may facilitate the development of adoptive cell therapies that work independently of antigen expression and of tissue of origin.


Asunto(s)
Neoplasias , Humanos , Ratones , Animales , Fluoresceína-5-Isotiocianato/metabolismo , Ligandos , Linfocitos T , Inmunoterapia Adoptiva
10.
Front Immunol ; 13: 886683, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35812387

RESUMEN

While immune checkpoint blockade results in durable responses for some patients, many others have not experienced such benefits. These treatments rely upon reinvigorating specific T cell-antigen interactions. However, it is often unknown what antigens are being recognized by T cells or how to potently induce antigen-specific responses in a broadly applicable manner. Here, we characterized the CD8+ T cell response to a murine model of melanoma following combination immunotherapy to determine the basis of tumor recognition. Sequencing of tumor-infiltrating T cells revealed a repertoire of highly homologous TCR sequences that were particularly expanded in treated mice and which recognized an antigen from an endogenous retrovirus. While vaccination against this peptide failed to raise a protective T cell response in vivo, engineered antigen mimotopes induced a significant expansion of CD8+ T cells cross-reactive to the original antigen. Vaccination with mimotopes resulted in killing of antigen-loaded cells in vivo yet showed modest survival benefit in a prophylactic vaccine paradigm. Together, this work demonstrates the identification of a dominant tumor-associated antigen and generation of mimotopes which can induce robust functional T cell responses that are cross-reactive to the endogenous antigen across multiple individuals.


Asunto(s)
Linfocitos T CD8-positivos , Melanoma , Animales , Antígenos de Neoplasias , Reacciones Cruzadas , Inmunoterapia , Melanoma/terapia , Ratones
11.
Nat Cancer ; 2(10): 1071-1085, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34738089

RESUMEN

Immune evasion is a hallmark of cancer, and therapies that restore immune surveillance have proven highly effective in cancers with high tumor mutation burden (TMB) (e.g., those with microsatellite instability (MSI)). Whether low TMB cancers, which are largely refractory to immunotherapy, harbor potentially immunogenic neoantigens remains unclear. Here, we show that tumors from all patients with microsatellite stable (MSS) colorectal cancer (CRC) express clonal predicted neoantigens despite low TMB. Unexpectedly, these neoantigens are broadly expressed at lower levels compared to those in MSI CRC. Using a versatile platform for modulating neoantigen expression in CRC organoids and transplantation into the distal colon of mice, we show that low expression precludes productive cross priming and drives immediate T cell dysfunction. Strikingly, experimental or therapeutic rescue of priming rendered T cells capable of controlling tumors with low neoantigen expression. These findings underscore a critical role of neoantigen expression level in immune evasion and therapy response.


Asunto(s)
Neoplasias Colorrectales , Linfocitos T , Animales , Antígenos de Neoplasias/genética , Neoplasias Colorrectales/genética , Humanos , Inmunoterapia , Ratones , Inestabilidad de Microsatélites
12.
ACS Appl Bio Mater ; 3(1): 180-185, 2020 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35019434

RESUMEN

Intracellular protein delivery using simple noncovalent carriers is an emerging field advancing the study of intracellular pathways and novel therapeutics. Here, we directly compare green fluorescent protein delivery using our recently reported protein transduction domain mimic (PTDM) to delivery with four commercially available amphiphilic macromolecular carriers in five diverse cell types. While most carriers succeeded only in serum-free conditions, the PTDM maintained robust delivery in complete media, even when tasked with antibody delivery into difficult-to-transfect neurons for the first time. The broad effectiveness of this reagent establishes PTDMs as a promising strategy for the delivery of biologics into such sensitive and challenging cell types.

13.
J Control Release ; 254: 131-136, 2017 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-28363520

RESUMEN

The impermeability of the plasma membrane towards large, hydrophilic biomolecules is a major obstacle in their use and development against intracellular targets. To overcome such limitations, protein transduction domains (PTDs) have been used as protein carriers, however they often require covalent fusion to the protein for efficient delivery. In an effort to develop more efficient and versatile biological vehicles, a series of PTD-inspired polyoxanorbornene-based synthetic mimics with identical chemical compositions but different hydrophobic/hydrophilic segregation were used to investigate the role of sequence segregation on protein binding and uptake into Jurkat T cells and HEK293Ts. This series was composed of a strongly segregated block copolymer, an intermediately segregated gradient copolymer, and a non-segregated homopolymer. Among the series, the block copolymer maximized both protein binding and translocation efficiencies, closely followed by the gradient copolymer, resulting in two protein transporter molecules more efficacious than currently commercially available agents. These two polymers were also used to deliver the biologically active Cre recombinase into a loxP-reporter T cell line. Since exogenous Cre must reach the nucleus and retain its activity to induce gene recombination, this in vitro experiment better exemplifies the broad applicability of this synthetic system. This study shows that increasing segregation between hydrophobic and cationic moieties in these polymeric mimics improves non-covalent protein delivery, providing crucial design parameters for the creation of more potent biological delivery agents for research and biomedical applications.


Asunto(s)
Portadores de Fármacos/química , Norbornanos/química , Péptidos/química , Polímeros/química , Proteínas/administración & dosificación , Proteínas/química , Transporte Biológico , Liberación de Fármacos , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Integrasas/metabolismo , Células Jurkat , Permeabilidad , Unión Proteica , Conformación Proteica , Dominios Proteicos , Proteínas/metabolismo , Recombinación Genética
14.
Polym Chem ; 7(48): 7514-7521, 2016 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-29093759

RESUMEN

The plasma membrane is a major obstacle in the development and use of biomacromolecules for intracellular therapeutic applications. Protein transduction domains (PTDs) have been used to overcome this barrier, but often require covalent conjugation to their cargo and can be time consuming to synthesize. Synthetic monomers can be designed to mimic the amino acid moieties in PTDs, and their resulting polymers provide a well-controlled platform to vary molecular composition for structure-activity relationship studies. In this paper, a series of polyoxanorbornene-based synthetic mimics, inspired by PTDs, with varying cationic and hydrophobic densities, and the nature of the hydrophobic chain and degree of polymerizations were investigated in vitro to determine their ability to non-covalently transport enhanced green fluorescent protein into HeLa cells, Jurkat T cells, and hTERT mesenchymal stem cells. Polymers with high charge density lead to efficient protein delivery. Similarly, the polymers with the highest hydrophobic content and density proved to be the most efficient at internalization. The observed improvements with increased hydrophobic length and content were consistent across all three cell types, suggesting that these architectural relationships are not cell type specific. However, Jurkat T cells showed distinct variation in uptake between polymers than with the other two cell types. These results provide important design parameters for more effective delivery of biomacromolecules for intracellular delivery applications.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA