Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cancer Sci ; 115(4): 1102-1113, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38287511

RESUMO

Worldwide prevalence of cervical cancer decreased significantly with the use of human papilloma virus (HPV)-targeted prophylactic vaccines. However, these multivalent antiviral vaccines are inert against established tumors, which leave patients with surgical ablative options possibly resulting in long-term reproductive complications and morbidity. In an attempt to bypass this unmet medical need, we designed a new E7 protein-based vaccine formulation using Accum™, a technology platform designed to promote endosome-to-cytosol escape as a means to enhance protein accumulation in target cells. Prophylactic vaccination of immunocompetent mice using the Accum-E7 vaccine (aE7) leads to complete protection from cervical cancer despite multiple challenges conducted with ascending C3.43 cellular doses (0.5-, 1.0-, and 2.0 × 106 cells). Moreover, the humoral response induced by aE7 was higher in magnitude compared with naked E7 protein vaccination and displayed potent inhibitory effects on C3.43 proliferation in vitro. When administered therapeutically to animals with pre-established C3.43 or Tal3 tumors, the vaccine-induced response synergized with multiple immune checkpoint blockers (anti-PD-1, anti-CTLA4, and anti-CD47) to effectively control tumor growth. Mechanistically, the observed therapeutic effect requires cross-presenting dendritic cells as well as CD8 T cells predominantly, with a non-negligible role played by both CD4+ and CD19+ lymphocytes. good laboratory practice (GLP) studies revealed that aE7 is immunogenic and well tolerated by immunocompetent mice with no observed adverse effects despite the use of a fourfold exceeding dose. In a nutshell, aE7 represents an ideal vaccine candidate for further clinical development as it uses a single engineered protein capable of exhibiting both prophylactic and therapeutic activity.


Assuntos
Vacinas Anticâncer , Proteínas Oncogênicas Virais , Infecções por Papillomavirus , Vacinas contra Papillomavirus , Neoplasias do Colo do Útero , Feminino , Humanos , Animais , Camundongos , Neoplasias do Colo do Útero/patologia , Proteínas E7 de Papillomavirus/metabolismo , Linfócitos T CD8-Positivos , Vacinação , Camundongos Endogâmicos C57BL , Infecções por Papillomavirus/prevenção & controle , Proteínas Oncogênicas Virais/genética
2.
J Transl Med ; 22(1): 532, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38831284

RESUMO

BACKGROUND: The Accum® platform was initially designed to accumulate biomedicines in target cells by inducing endosomal-to-cytosol escape. Interestingly however, the use of unconjugated Accum® was observed to trigger cell death in a variety of cancer cell lines; a property further exploited in the development of Accum®-based anti-cancer therapies. Despite the impressive pro-killing abilities of the parent molecule, some cancer cell lines exhibited resistance. This prompted us to test additional Accum® variants, which led to the identification of the AccuTOX® molecule. METHODS: A series of flow-cytometry and cell-based assays were used to assess the pro-killing properties of AccuTOX® along with its ability to trigger the production of reactive oxygen species (ROS), endosomal breaks and antigen presentation. RNA-seq was also conducted to pinpoint the most prominent processes modulated by AccuTOX® treatment in EL4 T-cell lymphoma. Finally, the therapeutic potency of intratumorally-injected AccuTOX® was evaluated in three different murine solid tumor models (EL4, E0771 and B16) both as a monotherapy or in combination with three immune-checkpoint inhibitors (ICI). RESULTS: In total, 7 Accum® variants were screened for their ability to induce complete cell death in 3 murine (EL4, B16 and E0771) and 3 human (MBA-MD-468, A549, and H460) cancer cell lines of different origins. The selected compound (hereafter refereed to as AccuTOX®) displayed an improved killing efficiency (~ 5.5 fold compared to the parental Accum®), while retaining its ability to trigger immunogenic cell death, ROS production, and endosomal breaks. Moreover, transcriptomic analysis revealed that low dose AccuTOX® enhances H2-Kb cell surface expression as well as antigen presentation in cancer cells. The net outcome culminates in impaired T-cell lymphoma, breast cancer and melanoma growth in vivo especially when combined with anti-CD47, anti-CTLA-4 or anti-PD-1 depending on the animal model. CONCLUSIONS: AccuTOX® exhibits enhanced cancer killing properties, retains all the innate characteristics displayed by the parental Accum® molecule, and synergizes with various ICI in controlling tumor growth. These observations will certainly pave the path to continue the clinical development of this lead compound against multiple solid tumor indications.


Assuntos
Sinergismo Farmacológico , Inibidores de Checkpoint Imunológico , Espécies Reativas de Oxigênio , Animais , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Linhagem Celular Tumoral , Humanos , Espécies Reativas de Oxigênio/metabolismo , Proliferação de Células/efeitos dos fármacos , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neoplasias/imunologia , Camundongos Endogâmicos C57BL , Feminino , Morte Celular/efeitos dos fármacos
3.
Cancer Sci ; 114(12): 4499-4510, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37776054

RESUMO

The Accum™ technology was initially designed to enhance the bioaccumulation of a given molecule in target cells. It does so by triggering endosomal membrane damages allowing endocytosed products to enter the cytosol, escaping the harsh environmental cues of the endosomal lumen. In an attempt to minimize manufacturing hurdles associated with Accum™ conjugation, we tested whether free Accum™ admixed with antigens could lead to outcomes similar to those obtained with conjugated products. Surprisingly, unconjugated Accum™ was found to promote cell death in vitro, an observation further confirmed on various murine tumor cell lines (EL4, CT-26, B16, and 4 T1). At the molecular level, unconjugated Accum™ triggers the production of reactive oxygen species and elicits immunogenic cell death while retaining its innate ability to cause endosomal damages. When administered as a monotherapy to animals with pre-established EL4 T-cell lymphoma, Accum™ controlled tumor growth in a dose-dependent manner, and its therapeutic effect relies on CD4 and CD8 T cells. Although unconjugated Accum™ synergizes with various immune checkpoint inhibitors (anti-CTLA4, anti-PD-1, or anti-CD47) at controlling tumor growth, its therapeutic potency could not be further enhanced when combined with all three tested immune checkpoint inhibitors at once due to its dependency on a specific dosing regimen. In sum, we report in this study an unprecedented new function for unconjugated Accum™ as a novel anticancer molecule. These results could pave the path for a new line of investigation aimed at exploring the pro-killing properties of additional Accum™ variants as a mean to develop second-generation anticancer therapeutics.


Assuntos
Inibidores de Checkpoint Imunológico , Linfoma de Células T , Animais , Camundongos , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral
4.
Molecules ; 27(12)2022 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-35744930

RESUMO

Compromised activity is a common impediment for biologics requiring endosome trafficking into target cells. In cancer cells, antibody-drug conjugates (ADCs) are trapped in endosomes or subsequently pumped extracellularly, leading to a reduction in intracellular accumulation. In subsets of dendritic cells (DCs), endosome-engulfed antigens face non-specific proteolysis and collateral damage to epitope immunogenicity before proteasomal processing and subsequent surface presentation. To bypass these shortcomings, we devised Accum™, a conjugable biotechnology harboring cholic acid (ChAc) and a nuclear localization signal (NLS) sequence for endosome escape and prompt nuclear targeting. Combined, these mechanisms culminate in enhanced intracellular accumulation and functionalization of coupled biologics. As proof-of-principle, we have biochemically characterized Accum, demonstrating its adaptability to ADCs or antigens in different cancer settings. Additionally, we have validated that endosome escape and nuclear routing are indispensable for effective intracellular accumulation and guaranteed target cell selectivity. Importantly, we have demonstrated that the unique mechanism of action of Accum translates into enhanced tumor cytotoxicity when coupled to ADCs, and durable therapeutic and prophylactic anti-cancer immunogenicity when coupled to tumor antigens. As more pre-clinical evidence accumulates, the adaptability, unique mechanism of action, and high therapeutic potency of Accum signal a promising transition into clinical investigations in the context of onco-immunotherapy.


Assuntos
Produtos Biológicos , Imunoconjugados , Antígenos de Neoplasias , Produtos Biológicos/farmacologia , Endossomos , Imunoconjugados/farmacologia , Imunoconjugados/uso terapêutico , Imunoterapia , Sinais de Localização Nuclear/química
5.
Bioconjug Chem ; 29(4): 1352-1363, 2018 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-29433309

RESUMO

Receptor-mediated internalization followed by trafficking and degradation of antibody-conjugates (ACs) via the endosomal-lysosomal pathway is the major mechanism for delivering molecular payloads inside target tumor cells. Although a mainstay for delivering payloads with clinically approved ACs in cancer treatment and imaging, tumor cells are often able to decrease intracellular payload concentrations and thereby reduce the effectiveness of the desired application. Thus, increasing payload intracellular accumulation has become a focus of attention for designing next-generation ACs. We developed a composite compound (ChAcNLS) that enables ACs to escape endosome entrapment and route to the nucleus resulting in the increased intracellular accumulation as an interleukin-5 receptor α-subunit (IL-5Rα)-targeted agent for muscle invasive bladder cancer (MIBC). We constructed 64Cu-A14-ChAcNLS, 64Cu-A14-NLS, and 64Cu-A14 and evaluated their performance by employing mechanistic studies for endosome escape coupled to nuclear routing and determining whether this delivery system results in improved 64Cu cellular accumulation. ACs consisting of ∼20 ChAcNLS or NLS moieties per 64Cu-A14 were prepared in good yield, high monomer content, and maintaining high affinity for IL-5Rα. Confocal microscopy analysis demonstrated ChAcNLS mediated efficient endosome escape and nuclear localization. 64Cu-A14-ChAcNLS increased 64Cu cellular accumulation in HT-1376 and HT-B9 cells relative to 64Cu-A14 and 64Cu-A14-NLS. In addition, we tested 64Cu-A14-ChAcNLS in vivo to evaluate its tissue distribution properties and, ultimately, tumor uptake and targeting. A model of human IL-5Rα MIBC was developed by implanting NOD/SCID mice with subcutaneous HT-1376 or HT-B9MIBC tumors, which grow containing high and low IL-5Rα-positive tumor cell densities, respectively. ACs were intravenously injected, and daily blood sampling, biodistribution at 48 and 96 h, and positron emission tomography (PET) at 24 and 48 h were performed. Region of interest (ROI) analysis was also performed on reconstructed PET images. Pharmacokinetic analysis and biodistribution studies showed that 64Cu-A14-ChAcNLS had faster clearance rates from the blood and healthy organs relative to 64Cu-A14. However, 64Cu-A14-ChAcNLS maintained comparable tumor accumulation relative to 64Cu-A14. This resulted in 64Cu-A14-ChAcNLS having superior tumor/normal tissue ratios at both 48 and 96 h biodistribution time points. Visualization of AC distribution by PET and ROI analysis confirmed that 64Cu-A14-ChAcNLS had improved targeting of MIBC tumor relative to 64Cu-A14. In addition, 64Cu-A14 modified with only NLS had poor tumor targeting. This was a result of poor tumor uptake due to extremely rapid clearance. Thus, the overall findings in this model of human IL-5Rα-positive MIBC describe an endosome escape-nuclear localization cholic-acid-linked peptide that substantially enhances AC cellular accumulation and tumor targeting.


Assuntos
Ácido Cólico/química , Ácido Cólico/farmacocinética , Imunoconjugados/química , Imunoconjugados/farmacocinética , Subunidade alfa de Receptor de Interleucina-5/análise , Neoplasias da Bexiga Urinária/diagnóstico por imagem , Animais , Linhagem Celular Tumoral , Ácido Cólico/administração & dosagem , Radioisótopos de Cobre/administração & dosagem , Radioisótopos de Cobre/química , Radioisótopos de Cobre/farmacocinética , Sistemas de Liberação de Medicamentos , Humanos , Imunoconjugados/administração & dosagem , Imunoconjugados/imunologia , Subunidade alfa de Receptor de Interleucina-5/imunologia , Camundongos Endogâmicos NOD , Camundongos SCID , Tomografia por Emissão de Pósitrons/métodos , Distribuição Tecidual , Neoplasias da Bexiga Urinária/imunologia , Neoplasias da Bexiga Urinária/terapia
6.
Bioorg Med Chem Lett ; 27(3): 443-446, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28049591

RESUMO

In vivo imaging of estrogen receptor (ER) densities in human breast cancer is a potential tool to stage disease, guide treatment protocols and follow-up on treatment outcome. Both positron emission tomography (PET) and fluorescence imaging have received ample attention to detect ligand-ER interaction. In this study we prepared BODIPY-estradiol conjugates using 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY) as fluorescent probe and estradiol derivatives as ligand and established their relative binding affinity (RBA) for the ERα. The synthesis of the conjugates involves attachment of a BODIPY moiety to the C17α-position of estradiol using Sonogashira or click reactions of iodo-BODIPY or aza-BODIPY with various 17α-ethynylestradiol (EE2) derivatives. The highest RBA for the ERα was observed with the EE2-BODIPY conjugate (7) featuring a linear eight carbon spacer chain. Cell uptake studies and in vivo imaging experiments in an ER-positive mouse tumor model are in progress.


Assuntos
Compostos de Boro/química , Meios de Contraste/síntese química , Receptor alfa de Estrogênio/metabolismo , Etinilestradiol/química , Neoplasias da Mama/diagnóstico por imagem , Química Click , Meios de Contraste/química , Meios de Contraste/metabolismo , Receptor alfa de Estrogênio/química , Feminino , Humanos , Concentração Inibidora 50 , Tomografia por Emissão de Pósitrons , Ligação Proteica , Espectrometria de Fluorescência
7.
Mol Pharm ; 13(6): 1915-26, 2016 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-27112376

RESUMO

The design of antibody-conjugates (ACs) for delivering molecules for targeted applications in humans has sufficiently progressed to demonstrate clinical efficacy in certain malignancies and reduced systemic toxicity that occurs with standard nontargeted therapies. One area that can advance clinical success for ACs will be to increase their intracellular accumulation. However, entrapment and degradation in the endosomal-lysosomal pathway, on which ACs are reliant for the depositing of their molecular payload inside target cells, leads to reduced intracellular accumulation. Innovative approaches that can manipulate this pathway may provide a strategy for increasing accumulation. We hypothesized that escape from entrapment inside the endosomal-lysosomal pathway and redirected trafficking to the nucleus could be an effective approach to increase intracellular AC accumulation in target cells. Cholic acid (ChAc) was coupled to the peptide CGYGPKKKRKVGG containing the nuclear localization sequence (NLS) from SV-40 large T-antigen, which is termed ChAcNLS. ChAcNLS was conjugated to the mAb 7G3 (7G3-ChAcNLS), which has nanomolar affinity for the cell-surface leukemic antigen interleukin-3 receptor-α (IL-3Rα). Our aim was to determine whether 7G3-ChAcNLS increased intracellular accumulation while retaining nanomolar affinity and IL-3Rα-positive cell selectivity. Competition ELISA and cell treatment assays were performed. Cell fractionation, confocal microscopy, flow cytometry, and Western blot techniques were used to determine the level of antibody accumulation inside cells and in corresponding nuclei. In addition, the radioisotope copper-64 ((64)Cu) was also utilized as a surrogate molecular cargo to evaluate nuclear and intracellular accumulation by radioactivity counting. 7G3-ChAcNLS effectively escaped endosome entrapment and degradation resulting in a unique intracellular distribution pattern. mAb modification with ChAcNLS maintained 7G3 nM affinity and produced high selectivity for IL-3Rα-positive cells. In contrast, 7G3 ACs with the ability to either escape endosome entrapment or traffic to the nucleus was not superior to 7G3-ChAcNLS for increasing intracellular accumulation. Transportation of (64)Cu when complexed to 7G3-ChAcNLS also resulted in increased nuclear and intracellular radioactivity accumulation. Thus, ChAcNLS is a novel mAb functionalizing technology that demonstrates its ability to increase AC intracellular accumulation in target cells through escaping endosome entrapment coupled to nuclear trafficking.


Assuntos
Anticorpos Monoclonais/farmacologia , Núcleo Celular/efeitos dos fármacos , Ácido Cólico/farmacocinética , Endossomos/efeitos dos fármacos , Imunoconjugados/farmacologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Endossomos/metabolismo , Humanos , Subunidade alfa de Receptor de Interleucina-3/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Sinais de Localização Nuclear/metabolismo , Peptídeos/metabolismo
8.
iScience ; 27(3): 109248, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38433914

RESUMO

Mesenchymal stromal cells (MSCs) have been modified via genetic or pharmacological engineering into potent antigen-presenting cells-like capable of priming responding CD8 T cells. In this study, our screening of a variant library of Accum molecule revealed a molecule (A1) capable of eliciting antigen cross-presentation properties in MSCs. A1-reprogrammed MSCs (ARM) exhibited improved soluble antigen uptake and processing. Our comprehensive analysis, encompassing cross-presentation assays and molecular profiling, among other cellular investigations, elucidated A1's impact on endosomal escape, reactive oxygen species production, and cytokine secretion. By evaluating ARM-based cellular vaccine in mouse models of lymphoma and melanoma, we observe significant therapeutic potency, particularly in allogeneic setting and in combination with anti-PD-1 immune checkpoint inhibitor. Overall, this study introduces a strong target for developing an antigen-adaptable vaccination platform, capable of synergizing with immune checkpoint blockers to trigger tumor regression, supporting further investigation of ARMs as an effective and versatile anti-cancer vaccine.

9.
Cell Rep Med ; 3(3): 100534, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35492876

RESUMO

The cross-presenting capacity of dendritic cells (DCs) can be limited by non-specific degradation during endosome maturation. To bypass this limitation, we present in this study a new Accum-based formulation designed to promote endosome-to-cytosol escape. Treatment of primary DCs with Accum linked to the xenoantigen ovalbumin (OVA) triggers endosomal damages and enhances protein processing. Despite multiple challenges using ascending doses of tumor cells, DC prophylactic vaccination results in complete protection due to increased levels of effector CD4 and CD8 T cells as well as high production of pro-inflammatory mediators. When combined with anti-PD-1, therapeutic vaccination using both syngeneic and allogeneic Accum-OVA-pulsed DCs triggers potent anti-tumoral responses. The net outcome culminates in increased CD11c, CD8, and NK infiltration along with a high CD8/Treg ratio. These highly favorable therapeutic effects highlight the promising potential of Accum as a distinct and potent technology platform suitable for the design of next generation cell cancer vaccines.


Assuntos
Vacinas Anticâncer , Células Dendríticas , Deriva e Deslocamento Antigênicos , Linfócitos T CD8-Positivos , Endossomos , Ovalbumina
10.
Artigo em Inglês | MEDLINE | ID: mdl-35457706

RESUMO

Climate change is a multidimensional issue that affects all aspects of society, including public health and human rights. Climate change is already severely impacting people's health and threatening people's guaranteed fundamental rights, including those to life, health, self-determination, and education, among others. Across geographical regions, population groups and communities who are already marginalized due to age, gender, ethnicity, income, and other socioeconomic factors, are those who are disproportionately affected by climate impacts despite having contributed the least to global emissions. Although scholars have been calling for a human rights-based approach and a health perspective to climate action, the literature looking at this multidisciplinary intersection is still nascent, and governments have yet to implement such intersectoral policies. This commentary begins to reflect on the relationship between climate change, human rights, and public health from the perspective of young people engaged in climate action and discourse at the national and international levels. It presents a way forward on what we, as youth climate advocates and researchers, believe is a priority to bring intersectoral integration of human rights and public health approaches to climate change to fruition. First, scholars and practitioners should examine and support youth-led climate interventions that tackle human rights and public health violations incurred by the climate crisis. Second, participatory approaches to climate change must be designed by working synergistically with climate-vulnerable groups, including children and young people, practitioners and scholars in public health and human rights sectors to holistically address the social, health, and environmental impacts of the climate crisis and root causes of injustice. Finally, we recommend more holistic data collection to better inform evidence-based climate policies that operationalize human rights and public health co-benefits.


Assuntos
Direitos Humanos , Saúde Pública , Adolescente , Criança , Mudança Climática , Humanos
11.
Biomed Opt Express ; 12(9): 5704-5719, 2021 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-34692210

RESUMO

Optical coherence tomography (OCT) was recently performed using a few-mode (FM) fiber to increase contrast or improve resolution using a sequential time-domain demultiplexing scheme isolating the different interferometric signals of the mode-coupled backscattered light. Here, we present an all-fiber FM-OCT system based on a parallel modal demultiplexing scheme exploiting a novel modally-specific photonic lantern (MSPL). The MSPL allows for maximal fringe visibility for each fiber propagation mode in an all-fiber assembly which provides the robustness required for clinical applications. The custom-built MSPL was designed for OCT at 930 nm and is wavelength-independent over the broad OCT spectrum. We further present a comprehensive coupling model for the interpretation of FM-OCT images using the first two propagation modes of a few-mode fiber, validate its predictions, and demonstrate the technique using in vitro microbead phantoms and ex vivo biological samples.

12.
Biochim Biophys Acta ; 1793(2): 335-45, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19014979

RESUMO

The observation that PrP is present in the cytosol of some neurons and non-neuronal cells and that the N-terminal signal peptide is slightly inefficient has brought speculations concerning a possible function of the protein in the cytosol. Here, we show that cells expressing a cytosolic form of PrP termed cyPrP display a large juxtanuclear cytoplasmic RNA organelle. Although cyPrP spontaneously forms aggresomes, we used several mutants to demonstrate that the assembly of this RNA organelle is independent from cyPrP aggregation. Components of the organelle fall into three classes: mRNAs; proteins, including the RNAseIII family polymerase Dicer, the decapping enzyme Dcp1a, the DEAD-box RNA helicase DDX6, and the small nuclear ribonucleoprotein-associated proteins SmB/B'/N; and non-coding RNAs, including rRNA 5S, tRNAs, U1 small nuclear RNA, and microRNAs. This composition is similar to RNA granules or chromatoid bodies from germ cells, or planarian stem cells and neurons, which are large ribonucleoprotein complexes predicted to function in RNA processing and posttranscriptional gene regulation. The domain of PrP encompassing residues 30 to 49 is essential for the formation of the RNA particle. Our findings confirm the intriguing relation between PrP and RNA in cells, and underscore an unexpected function for cytosolic PrP: assembling a large RNA processing center which we have termed PrP-RNP for PrP-induced ribonucleoprotein particle.


Assuntos
Grânulos Citoplasmáticos/metabolismo , Regulação da Expressão Gênica , Príons/metabolismo , RNA/metabolismo , Ribonucleoproteínas/metabolismo , Transcrição Gênica , Animais , Linhagem Celular Tumoral , Células Germinativas/metabolismo , Camundongos , Neurônios/metabolismo , Poro Nuclear/metabolismo , Planárias/citologia , Planárias/metabolismo , Ligação Proteica , RNA Ribossômico 18S/metabolismo , RNA Ribossômico 28S/metabolismo , RNA Nuclear Pequeno/metabolismo , RNA de Transferência/metabolismo , Ribonucleoproteínas/química , Células-Tronco/metabolismo , Vimentina/metabolismo
13.
Mol Ther Methods Clin Dev ; 19: 99-119, 2020 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-33024794

RESUMO

The next breakthrough for protein therapeutics is effective intracellular delivery and accumulation within target cells. Nuclear localization signal (NLS)-tagged therapeutics have been hindered by the lack of efficient nuclear localization due to endosome entrapment. Although development of strategies for tagging therapeutics with technologies capable of increased membrane penetration has resulted in proportional increased potency, nonspecific membrane penetration limits target specificity and, hence, widespread clinical success. There is a long-standing idea that nuclear localization of NLS-tagged agents occurs exclusively via classical nuclear transport. In the present study, we modified the antibody-drug conjugate trastuzumab-emtansine (T-DM1) with a classical NLS linked to cholic acid (cell accumulator [Accum]) that enables modified antibodies to escape endosome entrapment and increase nuclear localization efficiency without abrogating receptor targeting. In parallel, we developed a proteomics-based method to evaluate nuclear transport. Accum-modified T-DM1 significantly enhanced cytotoxic efficacy in the human epidermal growth factor receptor 2 (HER2)-positive SKBR3 breast cancer system. We discovered that efficacy was dependent on the nonclassical importin-7. Our evaluation reveals that when multiple classical NLS tagging occurs, cationic charge build-up as opposed to sequence dominates and becomes a substrate for importin-7. This study results in an effective target cell-specific NLS therapeutic and a general approach to guide future NLS-based development initiatives.

14.
Biochim Biophys Acta ; 1783(3): 479-91, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18023289

RESUMO

In mammalian cells, cytoplasmic protein aggregates generally coalesce to form aggresomal particles. Recent studies indicate that prion-infected cells produce prion protein (PrP) aggresomes, and that such aggregates may be present in the brain of infected mice. The molecular activity of PrP aggresomes has not been fully investigated. We report that PrP aggresomes initiate a cell stress response by activating the RNA-dependent protein kinase (PKR). Activated PKR phosphorylates the translation initiation factor eIF2alpha, resulting in protein synthesis shut-off. However, other components of the stress response, including the assembly of poly(A)+ RNA-containing stress granules and the synthesis of heat shock protein 70, are repressed. In situ hybridization experiments and affinity chromatography on oligo(dT)-cellulose showed that PrP aggresomes bind poly(A)+ RNA, and are therefore poly(A)+ ribonucleoprotein complexes. These findings support a model in which PrP aggresomes send neuronal cells into untimely demise by modifying the cell stress response, and by inducing the aggregation of poly(A)+ RNA.


Assuntos
Corpos de Inclusão Intranuclear/metabolismo , Príons/fisiologia , RNA Mensageiro/metabolismo , Ribonucleoproteínas/fisiologia , Estresse Fisiológico/metabolismo , eIF-2 Quinase/fisiologia , Animais , Células Cultivadas , Fator de Iniciação 2 em Eucariotos/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Células HeLa , Humanos , Corpos de Inclusão Intranuclear/fisiologia , Camundongos , Fosforilação , Príons/metabolismo , Biossíntese de Proteínas , Ribonucleoproteínas/metabolismo , Estresse Fisiológico/enzimologia , Estresse Fisiológico/etiologia
15.
BMC Cell Biol ; 9: 59, 2008 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-18937858

RESUMO

BACKGROUND: Aggresomes are juxtanuclear inclusion bodies that have been proposed to represent a general cellular response to misfolded proteins in mammalian cells. Yet, why aggresomes are not a pathological characteristic of protein misfolding diseases is unclear. Here, we investigate if a misfolded protein inevitably forms aggresomes in mammalian cells. RESULTS: We show that a cytoplasmic form of the prion protein may form aggresomes or dispersed aggregates in different cell lines. In contrast to aggresomes, the formation of dispersed aggregates is insensitive to histone deacetylase 6 inhibitors and does not result in cytoskeleton rearrangements. Modulation of expression levels or proteasome inhibitors does not alter the formation of dispersed aggregates. CONCLUSION: Our results establish that aggresomes are not obligatory products of protein misfolding in vivo.


Assuntos
Corpos de Inclusão/metabolismo , Príons/metabolismo , Dobramento de Proteína , Animais , Células COS , Chlorocebus aethiops , Citoplasma/metabolismo , Imunofluorescência , Células HeLa , Humanos , Hibridização in Situ Fluorescente , Camundongos , Células NIH 3T3 , Príons/análise , Príons/química
16.
J Vis Exp ; (133)2018 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-29578523

RESUMO

Antibody-conjugates (ACs) modified with virus-derived peptides are a potentially powerful class of tumor cell delivery agents for molecular payloads used in cancer treatment and imaging due to increased cellular accumulation over current ACs. During early AC in vitro development, fluorescence techniques and radioimmunoassays are sufficient for determining intracellular localization, accumulation efficiency, and target cell specificity. Currently, there is no consensus on standardized methods for preparing cells for evaluating AC intracellular accumulation and localization. The initial testing of ACs modified with virus-derived peptides is critical especially if several candidates have been constructed. Determining intracellular accumulation by fluorescence can be affected by background signal from ACs at the cell surface and complicate the interpretation of accumulation. For radioimmunoassays, typically treated cells are fractionated and the radioactivity in different cell compartments measured. However, cell lysis varies from cell to cell and often nuclear and cytoplasmic compartments are not adequately isolated. This can produce misleading data on payload delivery properties. The intravenous injection of radiolabeled virus-derived peptide-modified ACs in tumor bearing mice followed by radionuclide imaging is a powerful method for determining tumor targeting and payload delivery properties at the in vivo phase of development. However, this is a relatively recent advancement and few groups have evaluated virus-derived peptide-modified ACs in this manner. We describe the processing of treated cells to more accurately evaluate virus-derived peptide-modified AC accumulation when using confocal microscopy and radioimmunoassays. Specifically, a method for trypsinizing cells to remove cell surface bound ACs. We also provide a method for improving cellular fractionation. Lastly, this protocol provides an in vivo method using positron emission tomography (PET) for evaluating initial tumor targeting properties in tumor-bearing mice. We use the radioisotope 64Cu (t1/2 = 12.7 h) as an example payload in this protocol.


Assuntos
Anticorpos/imunologia , Imunoconjugados/uso terapêutico , Microscopia Confocal/métodos , Peptídeos/química , Tomografia por Emissão de Pósitrons/métodos , Animais , Linhagem Celular Tumoral , Radioisótopos de Cobre , Feminino , Humanos , Imunoconjugados/farmacologia , Camundongos
17.
Oncoimmunology ; 6(10): e1331195, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29123949

RESUMO

Despite the high interest and concern due to an increasing incidence and death rate, patients who develop muscle invasive bladder cancer (MIBC) have few options available. However, the past decade has produced many candidate bladder tumor-specific markers but further development of these markers is still needed for creating effective targeted medications to solve this urgent need. Interleukin-5 receptor α-subunit (IL-5Rα) has recently been reported to be involved in MIBC progression. Thus, we aimed to validate IL-5Rα as a target for antibody-conjugates to better manage patients with MIBC. Patients were recruited and their tumors were processed for IL-5Rα immunohistochemical analysis. NOD/SCID mice were also heterotopically implanted with the human MIBC HT-1376 and HT-B9 cell lines and established xenografts immunohistochemically evaluated for IL-5Rα and compared against patient tumors. Using the mAb A14, an antibody-drug conjugate (ADC) and a radiolabeled immunoconjugate (RIC) were developed by conjugating to vinblastine and to the positron emitter copper-64 (64Cu), respectively. As a proof-of-concept for ADC and RIC efficacy, in vitro cytotoxicity and in vivo positron emission tomography (PET) imaging in tumor-bearing mice were performed, respectively. In addition, as rapid internalization and accumulation are important components for effective antibody-conjugates, we evaluated these aspects in response to IL-5 and 64Cu-A14 treatments. Our findings suggest that although IL-5Rα protein expression is preferentially increased in MIBC, it is rapid IL-5Rα-mediated internalization allowing vinblastine-A14 to have cytotoxic activity and 64Cu-A14 to detect MIBC tumors in vivo. This is the first report to elucidate the potential of IL-5Rα as an attractive MIBC target for antibody-conjugate applications.

18.
Photochem Photobiol ; 92(4): 512-4, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27221219

RESUMO

There is considerable interest in the development of novel and more efficient delivery systems for improving the efficacy of photodynamic therapy (PDT). The authors in this highlighted issue describe the synthesis and the photobiological characterizations of two photosensitizer (PS) conjugates based on ß-carboline derivatives covalently conjugated to folic acid (FA) coupled to bovine serum albumin (BSA) as a carrier system specifically targeting cancer cells overexpressing FA receptor alpha (FRα). Accordingly, only the FA-BSA-ß-carboline conjugates are internalized specifically in FRα-positive cells and are proved to be phototoxic. On the other hand, albumin-ß-carboline conjugates without FA or ß-carboline derivatives alone are not internalized and nontoxic. This conjugate is among the first to produce a conjugate composed of a PS and FA molecules that are directly conjugated to BSA. In addition, the in vitro studies are the first evidence that directly conjugated FA-BSA can be used as carriers to selectively enhance cytotoxicity by PDT relative to unmodified PS or nontargeted BSA-PS. This strategy is a positive step forward for the covalent design and construction of a photodynamic nanomedicine for FR-positive tumors.


Assuntos
Ácido Fólico/química , Ácido Fólico/farmacologia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Soroalbumina Bovina/química , Soroalbumina Bovina/farmacologia , Carbolinas , Sistemas de Liberação de Medicamentos , Transportadores de Ácido Fólico , Humanos , Nanopartículas , Fotoquimioterapia
19.
Mol Neurodegener ; 8: 5, 2013 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-23339399

RESUMO

BACKGROUND: Aggregation of the α-Synuclein (α-Syn) protein, amyloid fibril formation and progressive neurodegeneration are the neuropathological hallmarks of Parkinson's Disease (PD). However, a detailed mechanism of α-Syn aggregation/fibrillogenesis and the exact nature of toxic oligomeric species produced during amyloid formation process are still unknown. RESULTS: In this study, the rates of α-Syn aggregation were compared for the recombinant wild-type (WT) α-Syn and a structurally relevant chimeric homologous protein containing an inducible Fv dimerizing domain (α-SynFv), capable to form dimers in the presence of a divalent ligand (AP20187). In the presence of AP20187, we report a rapid random coil into ß-sheet conformational transformation of α-SynFv within 24 h, whereas WT α-Syn showed 24 h delay to achieve ß-sheet structure after 48 h. Fluorescence ANS and ThT binding experiments demonstrate an accelerated oligomer/amyloid formation of dimerized α-SynFv, compared to the slower oligomerization and amyloidogenesis of WT α-Syn or α-SynFv without dimerizer AP20187. Both α-SynFv and α-Syn pre-fibrillar aggregates internalized cells and induced neurotoxicity when injected into the hippocampus of wild-type mice. These recombinant toxic aggregates further converted into non-toxic amyloids which were successfully amplified by protein misfolding cyclic amplification method, providing the first evidence for the in vitro propagation of synthetic α-Syn aggregates. CONCLUSIONS: Together, we show that dimerization is important for α-Syn conformational transition and aggregation. In addition, α-Syn dimerization can accelerate the formation of neurotoxic aggregates and amyloid fibrils which can be amplified in vitro. A detailed characterization of the mechanism of α-Syn aggregation/amyloidogenesis and toxicity is crucial to comprehend Parkinson's disease pathology at the molecular level.


Assuntos
Hipocampo/patologia , Multimerização Proteica , alfa-Sinucleína/química , alfa-Sinucleína/toxicidade , Animais , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doença de Parkinson/patologia , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA