Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Liposome Res ; : 1-26, 2024 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-38520185

RESUMO

Triple-negative breast Cancer (TNBC) is one of the deadliest types, making up about 20% of all breast cancers. Chemotherapy is the traditional manner of progressed TNBC treatment; however, it has a short-term result with a high reversibility pace. The lack of targeted treatment limited and person-dependent treatment options for those suffering from TNBC cautions to be the worst type of cancer among breast cancer patients. Consequently, appropriate treatment for this disease is considered a major clinical challenge. Therefore, various treatment methods have been developed to treat TNBC, among which chemotherapy is the most common and well-known approach recently studied. Although effective methods are chemotherapies, they are often accompanied by critical limitations, especially the lack of specific functionality. These methods lead to systematic toxicity and, ultimately, the expansion of multidrug-resistant (MDR) cancer cells. Therefore, finding novel and efficient techniques to enhance the targeting of TNBC treatment is an essential requirement. Liposomes have demonstrated that they are an effective method for drug delivery; however, among a large number of liposome-based drug delivery systems annually developed, a small number have just received authorization for clinical application. The new approaches to using liposomes target their structure with various ligands to increase therapeutic efficiency and diminish undesired side effects on various body tissues. The current study describes the most recent strategies and research associated with functionalizing the liposomes' structure with different ligands as targeted drug carriers in treating TNBCs in preclinical and clinical stages.

2.
Micromachines (Basel) ; 14(5)2023 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-37241658

RESUMO

Circulating tumor cells (CTCs) are cells that have been shed from tumors and circulate in the bloodstream. These cells can also be responsible for further metastases and the spread of cancer. Taking a closer look and analyzing CTCs through what has come to be known as "liquid biopsy" has immense potential to further researchers' understanding of cancer biology. However, CTCs are very sparse and are therefore difficult to detect and capture. To combat this issue, researchers have attempted to create devices, assays, and further techniques to successfully isolate CTCs for analysis. In this work, new and existing biosensing techniques for CTC isolation, detection, and release/detachment are discussed and compared to evaluate their efficacy, specificity, and cost. Here, we specifically aim to evaluate and identify the potential success of these techniques and devices in point-of-care (POC) settings.

3.
Pharmaceutics ; 15(2)2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36839837

RESUMO

The TRAIL (TNF-related apoptosis-inducing ligand) apoptotic pathway is extensively exploited in the development of targeted antitumor therapy due to TRAIL specificity towards its cognate receptors, namely death receptors DR4 and DR5. Although therapies targeting the TRAIL pathway have encountered many obstacles in attempts at clinical implementation for cancer treatment, the unique features of the TRAIL signaling pathway continue to attract the attention of researchers. Special attention is paid to the design of novel nanoscaled delivery systems, primarily aimed at increasing the valency of the ligand for improved death receptor clustering that enhances apoptotic signaling. Optionally, complex nanoformulations can allow the encapsulation of several therapeutic molecules for a combined synergistic effect, for example, chemotherapeutic agents or photosensitizers. Scaffolds for the developed nanodelivery systems are fabricated by a wide range of conventional clinically approved materials and innovative ones, including metals, carbon, lipids, polymers, nanogels, protein nanocages, virus-based nanoparticles, dendrimers, DNA origami nanostructures, and their complex combinations. Most nanotherapeutics targeting the TRAIL pathway are aimed at tumor therapy and theranostics. However, given the wide spectrum of action of TRAIL due to its natural role in immune system homeostasis, other therapeutic areas are also involved, such as liver fibrosis, rheumatoid arthritis, Alzheimer's disease, and inflammatory diseases caused by bacterial infections. This review summarizes the recent innovative developments in the design of nanodelivery systems modified with TRAIL pathway-targeting ligands.

4.
Nucleic Acid Ther ; 31(6): 404-416, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34468210

RESUMO

Ligand-targeted drug delivery (LTDD) has gained more attention in the field of nucleic acid therapeutics. To further elicit the potential of therapeutic oligonucleotides by means of LTDD, we newly developed (R)- and (S)-3-amino-1,2-propanediol (APD) manifold for ligand conjugation. N-acetylgalactosamine (GalNAc)/asialoglycoprotein receptor (ASGPr) system has been shown to be a powerful and robust paradigm of LTDD. Our novel APD-based GalNAc (GalNAcAPD) was shown to have intrinsic chemical instability that could play a role in better manipulation of active drug release. The APD manifold also enables facile production of conjugates through an on-support ligand cluster synthesis. We showed in a series of in vivo studies that while the knockdown activity of antisense oligonucleotides (ASOs) bearing 5'-GalNAcAPD was comparable to the conventional hydroxy-L-prolinol-linked GalNAc (GalNAcHP), 3'-GalNAcAPD elicited ASO activity by more than twice as much as the conventional 3'-GalNAcHP. This was ascribed partly to the GalNAcAPD's ideal susceptibility to nucleolytic digestion, which is expected to facilitate cytosolic internalization of ASO drugs. Moreover, an in vivo/ex vivo imaging study visualized the enhancement effect of monoantennary GalNAcAPD on liver localization of ASOs. This versatile manifold with chemical and biological instability would benefit therapeutic oligonucleotides that target both the liver and extrahepatic tissues.


Assuntos
Hepatócitos , Oligonucleotídeos , Acetilgalactosamina , Receptor de Asialoglicoproteína/genética , Ligantes , Oligonucleotídeos/genética
5.
Pharmaceutics ; 13(6)2021 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-34072682

RESUMO

The development of clinically relevant anti-microRNA antisense oligonucleotides (anti-miRNA ASOs) remains a major challenge. One promising configuration of anti-miRNA ASOs called "tiny LNA (tiny Locked Nucleic Acid)" is an unusually small (~8-mer), highly chemically modified anti-miRNA ASO with high activity and specificity. Within this platform, we achieved a great enhancement of the in vivo activity of miRNA-122-targeting tiny LNA by developing a series of N-acetylgalactosamine (GalNAc)-conjugated tiny LNAs. Specifically, the median effective dose (ED50) of the most potent construct, tL-5G3, was estimated to be ~12 nmol/kg, which is ~300-500 times more potent than the original unconjugated tiny LNA. Through in vivo/ex vivo imaging studies, we have confirmed that the major advantage of GalNAc over tiny LNAs can be ascribed to the improvement of their originally poor pharmacokinetics. We also showed that the GalNAc ligand should be introduced into its 5' terminus rather than its 3' end via a biolabile phosphodiester bond. This result suggests that tiny LNA can unexpectedly be recognized by endogenous nucleases and is required to be digested to liberate the parent tiny LNA at an appropriate time in the body. We believe that our strategy will pave the way for the clinical application of miRNA-targeting small ASO therapy.

6.
Curr Drug Deliv ; 18(8): 1094-1104, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33645481

RESUMO

Cardiovascular diseases cover various disorders like ischemic heart disease, hyperlipidemia, atherosclerosis, myocardial infarction, hypertension, etc. There are many synthetic drugs available for the treatment of cardiovascular therapy. However, they have several drawbacks like high dosing, toxicity, elevated blood potassium levels, low blood pressure, gastrointestinal issues, etc. To overcome these side effects of synthetic drugs, targeting the drug to the specific cardiac tissue is the best novel method in cardiovascular therapy. The highest targeting efficacy of ligand- based therapy with proper mechanisms and improved expandability provides a novel therapeutic strategy in cardiovascular diseases. Ligand therapy is more cost-effective compared to cell- based therapy. The surface area of protein is much larger than the orally bioavailable drug. Therefore, the targeting of various less active drug molecules to the particular ligand can be possible. The efficacy of ligands to induce cardiomyocytes proliferation has been ratified. The fact that ligand- based approaches are effective for cardiac transformation has been pointed out. Ligands interact with proteins in target cells, which are influenced by chemical signals. These various receptors selectively bind to biased ligands and energize the intracellular signaling pathway. The ligands can directly stabilize the active receptor conformations by a non-standard connective site. The key function of ligands is functional selectivity, which enhances the therapeutic efficacy and minimizes the side effects of drugs through the interpretation of signal transduction pathways. This review covers the role and effectiveness of novel ligands in cardiovascular disorders.


Assuntos
Doenças Cardiovasculares , Hipertensão , Hormônios Peptídicos , Receptores de Apelina , Doenças Cardiovasculares/tratamento farmacológico , Humanos , Ligantes
7.
Cell Prolif ; 53(9): e12880, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32707596

RESUMO

OBJECTIVES: To detect folate receptor (FR)-positive circulating tumour cells (FR+ CTCs) by using ligand-targeted polymerase chain reaction (LT-PCR) in periampullary cancer patients and to investigate the diagnostic value of FR+ CTCs in distinguishing pancreatic cancer (PC) from benign pancreatic disease. MATERIALS AND METHODS: CTCs were enriched from 3 mL of peripheral blood and portal vein blood by immunomagnetic depletion of leucocytes and were then detected by LT-PCR. The diagnostic performance of FR+ CTCs in PC was investigated by receiver-operating characteristic curve analysis. RESULTS: In total, 57 consecutive patients, including 46 patients with PC, five patients with non-pancreatic periampullary cancer (non-PC) and six patients with benign pancreatic diseases, were enrolled. FR+ CTC levels were significantly higher in patients with malignant diseases (PC and non-PC) than in patients with benign pancreatic diseases (P < .01). There was no notable difference in CTC levels between patients with PC and those with non-PC (P > .05). The combination of FR+ CTCs with carbohydrate antigen 19-9 (CA19-9) had better diagnostic efficiency than each of these two markers alone, with high sensitivity (97.8%) and specificity (83.3%). CONCLUSIONS: LT-PCR is feasible and reliable for detecting FR+ CTCs in patients with periampullary cancer. FR+ CTCs, especially when used in combination with CA19-9, have potential as a biomarker for the diagnosis of PC.


Assuntos
Receptores de Folato com Âncoras de GPI/análise , Células Neoplásicas Circulantes/patologia , Neoplasias Pancreáticas/diagnóstico , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos Glicosídicos Associados a Tumores/análise , Biomarcadores Tumorais/análise , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/patologia , Reação em Cadeia da Polimerase
8.
Biomolecules ; 10(6)2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32630411

RESUMO

Ligand-targeted toxins (LTTs) are bioengineered molecules which are composed of a targeting component linked to a toxin that induces cell death once the LTT binds its target. Bispecific targeting allows for the simultaneous targeting of two receptors. In this review, we mostly focus on the epidermal growth factor receptor (EGFR) as a target. We discuss the development and testing of a bispecific LTT targeting EGFR and urokinase-type plasminogen activator receptor (uPAR) as two attractive targets implicated in tumor growth and in the regulation of the tumor microvasculature in solid tumors. In vitro and mouse xenograft studies have shown that EGFR-targeted bispecific angiotoxin (eBAT) is effective against human solid tumors. Canine studies have shown that eBAT is both safe and effective against canine hemangiosarcoma, which is physiologically similar to human angiosarcoma. Finding the appropriate dosing strategy and sequencing of eBAT administration, in combination with other therapeutics, are among important factors for future directions. Together, the data indicate that eBAT targets cancer stem cells, it may have a role in inhibiting human tumor vasculature, and its bispecific conformation may have a role in reducing toxicity in comparative oncologic trials in dogs.


Assuntos
Anticorpos Biespecíficos/farmacologia , Antineoplásicos/farmacologia , Receptores ErbB/antagonistas & inibidores , Hemangiossarcoma/tratamento farmacológico , Receptores de Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Animais , Receptores ErbB/metabolismo , Hemangiossarcoma/metabolismo , Humanos , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo
9.
3 Biotech ; 10(4): 163, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32206497

RESUMO

Liposomes are very useful biocompatible tools used in diverse scientific disciplines, employed for the vehiculation and delivery of lipophilic, ampiphilic or hydrophilic compounds. Liposomes have gained the importance as drug carriers, as the drugs alone have limited targets, higher toxicity and develop resistance when used in higher doses. Conventional liposomes suffer from several drawbacks like encapsulation inefficiencies and partially controlled particle size. The surface chemistry of liposome technology started from simple conventional vesicles to second generation liposomes by modulating their lipid composition and surface with different ligands. Introduction of polyethylene glycol to lipid anchor was the first innovative strategy which increased circulation time, delayed clearance and opsonin resistance. PEGylated liposomes have been found to possess higher drug loading capacity up to 90% or more and some drugs like CPX-1 encapsuled in such liposomes have increased the disease control up to 73% patients suffering from colorectal cancer. The surface of liposomes have been further liganded with small molecules, vitamins, carbohydrates, peptides, proteins, antibodies, aptamers and enzymes. These advanced liposomes exhibit greater solubility, higher stability, long-circulating time and specific drug targeting properties. The immense utility and demand of surface modified liposomes in different areas have led their way to the modern market. In addition to this, the multi-drug carrier approach of targeted liposomes is an innovative method to overcome drug resistance while treating ceratin tumors. Presently, several second-generation liposomal formulations of different anticancer drugs are at various stages of clinical trials. This review article summarizes briefly the preparation of liposomes, strategies of disease targeting and exclusively the surface modifications with different entities and their clinical applications especially as drug delivery system.

10.
Acta Biomater ; 102: 13-34, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31759124

RESUMO

Among various nanoparticles, superparamagnetic iron oxide nanoparticles (SPIONs) have been increasingly studied for their excellent superparamagnetism, magnetic heating properties, and enhanced magnetic resonance imaging (MRI). The conjugation of SPIONs with drugs to obtain delivery nanosystems has several advantages including magnetic targeted functionalization, in vivo imaging, magnetic thermotherapy, and combined delivery of anticancer agents. To further increase the targeting efficiency of drugs through a delivery nanosystem based on SPIONs, additional targeting moieties including transferrin, antibodies, aptamers, hyaluronic acid, folate, and targeting peptides are coated onto the surface of SPIONs. Therefore, this review summarizes the latest progresses in the conjugation of targeting molecules and drug delivery nanosystems based on SPIONs, especially focusing on their performances to develop efficient targeted drug delivery systems for tumor therapy. STATEMENT OF SIGNIFICANCE: Some magnetic nanoparticle-based nanocarriers loaded with drugs were evaluated in patients and did not produce convincing results, leading to termination of clinical development in phase II/III. An alternative strategy for drug delivery systems based on SPIONs is the conjugation of these systems with targeting segments such as transferrin, antibodies, aptamers, hyaluronic acid, folate, and targeting peptides. These targeting moieties can be recognized by specific integrin/receptors that are overexpressed specifically on the tumor cell surface, resulting in minimizing dosage and reducing off-target effects. This review focuses on magnetic nanoparticle-based nonviral drug delivery systems with targeting moieties to deliver anticancer drugs, with an aim to provide suggestions on the development of SPIONs through discussion.


Assuntos
Antineoplásicos/uso terapêutico , Portadores de Fármacos/química , Nanopartículas Magnéticas de Óxido de Ferro/química , Neoplasias/tratamento farmacológico , Peptídeos/química , Animais , Ácido Fólico/química , Humanos , Ácido Hialurônico/química , Neoplasias/diagnóstico por imagem
11.
Mol Imaging Biol ; 22(2): 377-383, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31292915

RESUMO

PURPOSE: Neuroendocrine tumors (NETs) have reasonably high 5-year survival rates when diagnosed at an early stage but are significantly more lethal when discovered only after metastasis. Although several imaging modalities such as computed tomography (CT), positron emission tomography, and magnetic resonance imaging can detect neuroendocrine tumors, their high false positive rates suggest that more specific diagnostic tests are required. Targeted imaging agents such as Octreoscan® have met some of this need for improved specificity, but their inability to image poorly differentiated NETs suggests that improved NET imaging agents are still needed. Because neurokinin 1 receptors (NK1Rs) are widely over-expressed in neuroendocrine tumors, but show limited expression in healthy tissues, we have undertaken to develop an NK1R-targeted imaging agent for improved diagnosis and staging of neuroendocrine tumors. PROCEDURE: A small molecule NK1R antagonist was conjugated via a flexible spacer to a Tc-99m chelating peptide. After complexation with Tc-99m, binding of the conjugate to human embryonic kidney (HEK293) cells transfected with the human NK1R was evaluated as a function of radioimaging agent concentration. In vivo imaging of HEK293-NK1R tumor xenografts in mice was also performed by single-photon emission computed tomography/computed tomography (γ-SPECT/CT), and the distribution of the conjugate in various tissues was quantified by tissue resection and γ-counting. RESULTS: NK1R-targeted Tc-99m-based radioimaging agent displayed excellent affinity (Kd = 16.8 nM) and specificity for HEK293-NK1R tumor xenograft. SPECT/CT analysis of tumor-bearing mice demonstrated significant tumor uptake and high tumor to background ratio as early as 2 h post injection. CONCLUSION: The excellent tumor contrast afforded by our NK1R-targeted radioimaging agent exhibits properties that could improve early diagnosis and staging of many neuroendocrine tumors.


Assuntos
Tumores Neuroendócrinos/diagnóstico por imagem , Receptores da Neurocinina-1/química , Tecnécio/química , Animais , Quelantes/química , Reações Falso-Positivas , Feminino , Células HEK293 , Humanos , Ligantes , Camundongos , Camundongos Nus , Transplante de Neoplasias , Peptídeos/química , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único , Somatostatina/análogos & derivados , Tomografia Computadorizada por Raios X
12.
Nucleosides Nucleotides Nucleic Acids ; 39(1-3): 109-118, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31617782

RESUMO

The asialoglycoprotein receptor (ASGPr) and N-acetylgalactosamine (GalNAc) is one of the most reliable receptor-ligand combinations for delivering antisense oligonucleotides (ASOs) to the liver. Here, we show that a modular GalNAc conjugation strategy allows us to reinforce the activity of the parent, naked 2',4'-BNA/LNA gapmer targeting apolipoprotein B. The conjugation partly reduced a possible hepatotoxicity of the parent ASO. The structure-activity study revealed the significance of the metabolic susceptibility of the GalNAc moiety to nucleolytic cleavage that results in exposure of the parent gapmer. The broad usefulness of our delivery strategy of ASOs to the liver has been demonstrated.


Assuntos
Acetilgalactosamina , Oligonucleotídeos Antissenso , Acetilgalactosamina/química , Animais , Receptor de Asialoglicoproteína , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Humanos , Ligantes , Fígado , Masculino , Camundongos , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/química , Ligação Proteica
13.
Methods Mol Biol ; 2054: 283-294, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31482462

RESUMO

A series of ligand-targeted nanosystems have been rapidly exploited to selectively deliver drug molecules to desired cell populations. The conjugation of protein ligands to the nanoparticle (NP) surface endows nanovehicles with active targeting properties. However, the nonspecific covalent coupling of protein ligands to nanocarriers may compromise the protein targeting due to the uncontrolled ligand orientation as well as the decline in ligand activity during linkage process. With this regard, biomimetic synthetic strategies are employed for the preparation of genetically engineered nanovesicles (GNV) from cellular plasma membrane with targeting moieties on the surface in a ligand-oriented manner. Herein, we introduce the biomimetic synthetic strategy and procedures for GNV preparation. This chapter may guide readers to design analogous NPs for cell-specific targeting by displaying particular protein probes (e.g., antibody, nanobody, and single-chain antibody) on the surface of GNVs.


Assuntos
Antineoplásicos/administração & dosagem , Engenharia Genética/métodos , Nanopartículas/química , Neoplasias/terapia , Nanomedicina Teranóstica/métodos , Animais , Antineoplásicos/farmacocinética , Materiais Biomiméticos/síntese química , Linhagem Celular Tumoral , Membrana Celular/genética , Terapia Combinada/métodos , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Composição de Medicamentos/métodos , Exossomos/genética , Humanos , Hipertermia Induzida/métodos , Lipossomos , Camundongos , Neoplasias/diagnóstico por imagem , Neoplasias/patologia , Técnicas Fotoacústicas , Fotoquimioterapia/métodos , Anticorpos de Cadeia Única/administração & dosagem , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Control Release ; 311-312: 190-200, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31473250

RESUMO

Despite ligand-targeted liposomes long garnering interest as drug delivery vehicles for cancer therapeutics, inconsistency in successful outcomes have hindered their translation into the clinic. This is in part due to discrepancies between in vitro design evaluations and final in vivo outcomes. By employing a multifaceted synthetic strategy to prepare peptide-targeted nanoparticles of high purity, reproducibility, and with precisely controlled quantity of functionalities, we systematically evaluated the individual roles that peptide-linker length, peptide hydrophilicity, peptide density, and nanoparticle size play on cancer cell uptake and tumor targeting both in vitro and in vivo, and how the results correlated and contrasted. These parameters were analyzed using a VLA-4-targeted liposome system in a multiple myeloma mouse xenograft model to evaluate in vivo biodistribution and tumor cell uptake. The results showed that using in vitro models to optimize targeted-nanoparticles for maximum cellular uptake was helpful in narrowing down the particle characteristics. However, in vitro optimization fell short of achieving enhanced results in animal models, rather had negative consequences for in vivo targeting. This outcome is not surprising considering that the receptor being targeted is also present on healthy lymphocytes and increasing targeting peptide valency on particle surfaces results in an increase in non-selective, off-target binding to healthy cells. Hence, further optimization using in vivo models was absolutely necessary, through which we were able to increase the uptake of peptide-targeted liposomes by cancerous cells overexpressing VLA-4 to 15-fold over that of non-targeted liposomes in vivo. The results highlighted the importance of creating a comprehensive understanding of the effect of each liposome design parameter on multifactorial biological endpoints including both in vitro and in vivo in determining the therapeutic potential of peptide-targeted liposomes.


Assuntos
Integrina alfa4beta1/antagonistas & inibidores , Mieloma Múltiplo/tratamento farmacológico , Nanopartículas/administração & dosagem , Peptídeos/administração & dosagem , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Lipossomos , Camundongos SCID , Mieloma Múltiplo/metabolismo , Nanopartículas/química , Peptídeos/química
15.
Curr Protoc Nucleic Acid Chem ; 78(1): e99, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31529782

RESUMO

Ligand-targeted drug delivery (LTDD) has emerged as an attractive option in the field of oligonucleotide drugs following the great success of N-acetylgalactosamine (GalNAc)-conjugated siRNA and antisense oligonucleotides. GalNAc is a well-known ligand of the asialoglycoprotein receptor (ASGPR), and is classified as a C-type lectin associated with the metabolism of desialylated glycoproteins. This article describes the synthesis of a non-nucleosidic monovalent GalNAc phosphoramidite-a useful reagent for facilitating the conjugation of GalNAc epitopes into oligonucleotides using DNA synthesizers-together with some important caveats. The monomeric GalNAc consists of three parts: (1) a GalNAc moiety, (2) a linker moiety, and (3) a trans-4-hydroxyprolinol (tHP) branch point. The GalNAc moiety and the tHP moiety are coupled via a condensation reaction to prepare the monovalent GalNAc phosphoramidite. © 2019 by John Wiley & Sons, Inc. Basic Protocol 1: Synthesis of N-acetylgalactosamine ligand Basic Protocol 2: Preparation of trans-4-hydroxyprolinol building block Basic Protocol 3: Preparation of GalNAc phosphoramidite.


Assuntos
Acetilgalactosamina/química , Fígado/efeitos dos fármacos , Oligonucleotídeos/farmacologia , Compostos Organofosforados/química , Sistemas de Liberação de Medicamentos , Ligantes , Oligonucleotídeos/administração & dosagem , Oligonucleotídeos/química
16.
Curr Med Chem ; 26(3): 396-426, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-28820071

RESUMO

Monoclonal antibodies (mAbs) are an important class of therapeutic agents approved for the therapy of many types of malignancies. However, in certain cases applications of conventional mAbs have several limitations in anticancer immunotherapy. These limitations include insufficient efficacy and adverse effects. The antigen-binding fragments of antibodies have a considerable potential to overcome the disadvantages of conventional mAbs, such as poor penetration into solid tumors and Fc-mediated bystander activation of the immune system. Fragments of antibodies retain antigen specificity and part of functional properties of conventional mAbs and at the same time have much better penetration into the tumors and a greatly reduced level of adverse effects. Recent advantages in antibody engineering allowed to produce different types of antibody fragments with improved structure and properties for efficient elimination of tumor cells. These molecules opened up new perspectives for anticancer therapy. Here, we will overview the structural features of the various types of antibody fragments and their applications for anticancer therapy as separate molecules and as part of complex conjugates or structures. Mechanisms of antitumor action of antibody fragments as well as their advantages and disadvantages for clinical application will be discussed in this review.


Assuntos
Produtos Biológicos/uso terapêutico , Fragmentos de Imunoglobulinas/uso terapêutico , Neoplasias/tratamento farmacológico , Antígenos/imunologia , Humanos , Fragmentos de Imunoglobulinas/imunologia
17.
Mycopathologia ; 184(1): 181-185, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30101407

RESUMO

The widespread use of T lymphocyte-associated antigen-4 (CTLA-4) and programmed death (PD)-1 and PD ligand-1 (PDL1)-targeted agents in cancer patients as immunotherapy has raised some issues on their safety profile. Regarding infectious complications, it has emerged that these compounds do not intrinsically increase susceptibility to opportunistic infections, which mainly correlate with the co-administration of systemic immunosuppressive therapy (high-dose corticosteroids and anti-tumor necrosis factors inhibitors) to cure immune-related adverse events (colitis, hepatitis, pneumonitis and pancreatitis), well-known complications of these targeted drugs. These observations lead experts' opinion to suggest primary anti-Pneumocystis prophylaxis in patients undergoing CTLA-4 and PD-1/PDL1 agents who will receive prednisone 20 mg daily for ≥ 4 weeks. Few data on invasive fungal infections in this context are available. We report here a case of probable invasive pulmonary aspergillosis (p-IPA) complicating first-line immunotherapy with pembrolizumab for metastatic lung cancer that was further aggravated by multidrug-resistant Pseudomonas aeruginosa superinfection of fungal cavities; the patient received concurrent systemic corticosteroid therapy as anti-edema treatment for cerebral metastases. Reviewing literature about Aspergillus diseases in subjects receiving CTLA-4 and PD-1 and PDL1-targeted agents, we found three cases of invasive aspergillosis and one case of exacerbation of chronic progressive pulmonary aspergillosis after nivolumab treatment; to the best of our knowledge, this is the first report of p-IPA complicating pembrolizumab immunotherapy. Briefly, in this new setting of biological/targeted drugs, waiting for growing clinical experience, we recommend a high level of alertness in diagnosing any infectious complications.


Assuntos
Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/secundário , Anticorpos Monoclonais Humanizados/administração & dosagem , Antineoplásicos Imunológicos/administração & dosagem , Aspergilose Pulmonar Invasiva/diagnóstico , Infecções por Pseudomonas/diagnóstico , Adenocarcinoma de Pulmão/complicações , Corticosteroides/administração & dosagem , Corticosteroides/efeitos adversos , Anticorpos Monoclonais Humanizados/efeitos adversos , Antineoplásicos Imunológicos/efeitos adversos , Farmacorresistência Bacteriana Múltipla , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Feminino , Humanos , Imunossupressores/administração & dosagem , Imunossupressores/efeitos adversos , Aspergilose Pulmonar Invasiva/complicações , Aspergilose Pulmonar Invasiva/patologia , Pessoa de Meia-Idade , Infecções por Pseudomonas/complicações , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/isolamento & purificação
18.
Curr Protoc Chem Biol ; 10(4): e49, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30212611

RESUMO

The development of small molecule ligand-targeted therapeutics is currently of paramount importance for treatment of cancer due to their potential to reduce system toxicity and increase potency of a delivered chemotherapeutic drug. The main aim of a targeted drug-delivery technique is to release the drug cargo selectively into tumor tissues, avoiding off-site toxicity to healthy tissues and organs during chemotherapy. In this strategy, a chemotherapeutic drug is conjugated to a homing ligand, which has high affinity for proteins over-expressed on cancer cells, via a peptide linker and a self-immolative segment that facilitates intracellular release of drug cargo. During development of targeted drug conjugates, preclinical evaluation in tumor models of small animals like mice adds valuable data on the clinical performance of the drug. This article contains a set of protocols for implantation of tumor, determination of optimum dosage required for effective treatment, and estimation of maximum tolerated dose required for any visible side effects during treatment of cancer in tumor models of mice. © 2018 by John Wiley & Sons, Inc.


Assuntos
Antineoplásicos/farmacologia , Peptídeos/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Ligantes , Masculino , Camundongos , Camundongos Nus , Células PC-3 , Peptídeos/síntese química , Peptídeos/química , Neoplasias da Próstata/patologia , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Front Pharmacol ; 9: 787, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30072901

RESUMO

Since Human Genome Project (HGP) revealed the heterogeneity of individuals, precision medicine that proposes the customized healthcare has become an intractable and hot research. Meanwhile, as the Precision Medicine Initiative launched, precision drug design which aims at maximizing therapeutic effects while minimizing undesired side effects for an individual patient has entered a new stage. One of the key strategies of precision drug design is target based drug design. Once a key pathogenic target is identified, rational drug design which constitutes the major part of precision drug design can be performed. Examples of rational drug design on novel druggable targets and protein-protein interaction surfaces are summarized in this review. Besides, various kinds of computational modeling and simulation approaches increasingly benefit for the drug discovery progress. Molecular dynamic simulation, drug target prediction and in silico clinical trials are discussed. Moreover, due to the powerful ability in handling high-dimensional data and complex system, deep learning has efficiently promoted the applications of artificial intelligence in drug discovery and design. In this review, deep learning methods that tailor to precision drug design are carefully discussed. When a drug molecule is discovered, the development of specific targeted drug delivery system becomes another key aspect of precision drug design. Therefore, state-of-the-art techniques of drug delivery system including antibody-drug conjugates (ADCs), and ligand-targeted conjugates are also included in this review.

20.
Adv Healthc Mater ; 7(20): e1800269, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29956504

RESUMO

Ligand-targeted nanosystems have the potential to realize site-specific tumor therapy and alleviate unwanted side effects of many chemotherapeutic agents, and one of the most key issues seems to be the construction of an effective nanocarrier. Based on different processes of phage display techniques, 38 cell-binding peptides and 32 cell-internalizing peptides are discovered. Four of these ligand peptides [FIPFDPMSMRWE (FIP), NASSFPTNSRWA (NAS), GLHTSATNLYLH (GLH), and ALAVAPSRWWNE (ALA), respectively] exhibit high affinity to MCF7 human breast cancer cells. Among them, NAS and ALA are reported for the first time, whose affinities are 20.6 and 76.3 times that of the random peptide control, respectively. Both NAS and ALA modifications to doxorubicin-loaded lipid nanosytems [LP(DOX)] show stronger tumor inhibition, longer animal survival time, and less body weight loss, compared to unmodified or control peptide modified nanosystems, on an MCF7 tumor-bearing mouse model. In conclusion, the cell-binding peptide NAS and cell-internalizing peptide ALA can be used for ligand-targeted delivery of antitumor drugs. It seems that the in vivo antitumor effect of these ligand-targeted nanosystems is closely related to their ligand-cell affinity, but fairly tolerant of the ligand types.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Peptídeos/química , Animais , Doxorrubicina/química , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Lipossomos/química , Células MCF-7 , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA