Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Small ; : e2302931, 2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37525558

ABSTRACT

Combinations of chemotherapeutic agents comprise a clinically feasible approach to combat cancers that possess resistance to treatment. Type II endometrial cancer is typically associated with poor outcomes and the emergence of chemoresistance. To overcome this challenge, a combination therapy is developed comprising a novel ciprofloxacin derivative-loaded PEGylated polymeric nanoparticles (CIP2b-NPs) and paclitaxel (PTX) against human type-II endometrial cancer (Hec50co with loss of function p53). Cytotoxicity studies reveal strong synergy between CIP2b and PTX against Hec50co, and this is associated with a significant reduction in the IC50 of PTX and increased G2/M arrest. Upon formulation of CIP2b into PEGylated polymeric nanoparticles, tumor accumulation of CIP2b is significantly improved compared to its soluble counterpart; thus, enhancing the overall antitumor activity of CIP2b when co-administered with PTX. In addition, the co-delivery of CIP2b-NPs with paclitaxel results in a significant reduction in tumor progression. Histological examination of vital organs and blood chemistry was normal, confirming the absence of any apparent off-target toxicity. Thus, in a mouse model of human endometrial cancer, the combination of CIP2b-NPs and PTX exhibits superior therapeutic activity in targeting human type-II endometrial cancer.

2.
Int J Mol Sci ; 23(4)2022 Feb 17.
Article in English | MEDLINE | ID: mdl-35216330

ABSTRACT

Selenium has been extensively evaluated clinically as a chemopreventive agent with variable results depending on the type and dose of selenium used. Selenium species are now being therapeutically evaluated as modulators of drug responses rather than as directly cytotoxic agents. In addition, recent data suggest an association between selenium base-line levels in blood and survival of patients with COVID-19. The major focus of this mini review was to summarize: the pathways of selenium metabolism; the results of selenium-based chemopreventive clinical trials; the potential for using selenium metabolites as therapeutic modulators of drug responses in cancer (clear-cell renal-cell carcinoma (ccRCC) in particular); and selenium usage alone or in combination with vaccines in the treatment of patients with COVID-19. Critical therapeutic targets and the potential role of different selenium species, doses, and schedules are discussed.


Subject(s)
COVID-19 Drug Treatment , Neoplasms/drug therapy , Selenium/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , COVID-19/virology , DNA Repair/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , NF-E2-Related Factor 2/chemistry , NF-E2-Related Factor 2/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/isolation & purification , Selenium/chemistry , Selenium/metabolism , Selenium/pharmacology
3.
AAPS PharmSciTech ; 22(2): 71, 2021 Feb 11.
Article in English | MEDLINE | ID: mdl-33575970

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive type of malignant brain tumor. Current FDA-approved treatments include surgical resection, radiation, and chemotherapy, while hyperthermia, immunotherapy, and most relevantly, nanoparticle (NP)-mediated delivery systems or combinations thereof have shown promise in preclinical studies. Drug-carrying NPs are a promising approach to brain delivery as a result of their potential to facilitate the crossing of the blood-brain barrier (BBB) via two main types of transcytosis mechanisms: adsorptive-mediated transcytosis (AMT) and receptor-mediated transcytosis (RMT). Their ability to accumulate in the brain can thus provide local sustained release of tumoricidal drugs at or near the site of GBM tumors. NP-based drug delivery has the potential to significantly reduce drug-related toxicity, increase specificity, and consequently improve the lifespan and quality of life of patients with GBM. Due to significant advances in the understanding of the molecular etiology and pathology of GBM, the efficacy of drugs loaded into vectors targeting this disease has increased in both preclinical and clinical settings. Multitargeting NPs, such as those incorporating multiple specific targeting ligands, are an innovative technology that can lead to decreased off-target effects while simultaneously having increased accumulation and action specifically at the tumor site. Targeting ligands can include antibodies, or fragments thereof, and peptides or small molecules, which can result in a more controlled drug delivery system compared to conventional drug treatments. This review focuses on GBM treatment strategies, summarizing current options and providing a detailed account of preclinical findings with prospective NP-based approaches aimed at improving tumor targeting and enhancing therapeutic outcomes for GBM patients.


Subject(s)
Brain Neoplasms/drug therapy , Drug Delivery Systems , Glioblastoma/drug therapy , Nanoparticles/chemistry , Glioblastoma/pathology , Humans , Prospective Studies
4.
Biomacromolecules ; 21(2): 534-544, 2020 02 10.
Article in English | MEDLINE | ID: mdl-31895553

ABSTRACT

Leptospirosis is a debilitating infectious disease that detrimentally affects both animals and humans; therefore, disease prevention has become a high priority to avoid high incidence rates of disease in the herd and break the transmission cycle to humans. Thus, there remains an important unmet need for a prophylactic vaccine that can provide long-term immunity against leptospirosis in cattle. Herein, a novel vaccine formulation was developed where poly(diaminosulfide) polymer was employed to fabricate microparticles encapsulating the antigen of Leptospira borgpetersenii serovar Hardjo strain HB15B203 (L203-PNSN). A prime-boost vaccination with a L203-PNSN microparticle formulation increased the population of L203-specific CD3+ T cells and CD21+ B cells to levels that were significantly higher than those of cattle vaccinated with L203-AlOH or the vehicle control (empty PNSN microparticles and blank AlOH). In addition, L203-PNSN was demonstrated to stimulate durable humoral immune responses as evidenced by the increases in the antibody serum titers following the vaccination. It was also found that cattle vaccinated with L203-PNSN produced higher macroscopic agglutinating titers than cattle in other groups. Thus, it can be concluded that L203-PNSN is a novel first-in-class microparticle-based Leptospira vaccine that represents a powerful platform with the potential to serve as a prophylactic vaccine against leptospiral infection in cattle.


Subject(s)
Antigens, Bacterial/administration & dosage , Bacterial Vaccines/administration & dosage , Leptospira/immunology , Leptospirosis/prevention & control , Microplastics/chemistry , Animals , Bacterial Vaccines/immunology , Cattle , Cattle Diseases/microbiology , Cattle Diseases/prevention & control , Drug Delivery Systems/methods , Enzyme-Linked Immunosorbent Assay , Immunity, Humoral , Immunization, Secondary , Leptospirosis/immunology , Leptospirosis/veterinary , Male , Microplastics/chemical synthesis , Polymers/chemistry , T-Lymphocytes/immunology
5.
J Pharmacol Exp Ther ; 370(3): 855-863, 2019 09.
Article in English | MEDLINE | ID: mdl-30361239

ABSTRACT

Many factors affect vaccine efficacy. One of the most salient is the frequency and intervals of vaccine administration. In this study, we assessed the vaccine administration modality for a recently reported polyanhydride-based vaccine formulation, shown to generate antitumor activity. Polyanhydride particles encapsulating ovalbumin (OVA) were prepared using a double-emulsion technique and subcutaneously delivered to mice either as a single-dose or as prime-boost vaccine regimens in which two different time intervals between prime and boost were assessed (7 or 21 days). This was followed by measurement of cellular and humoral immune responses, and subsequent challenge of the mice with a lethal dose of E.G7-OVA cells to evaluate tumor protection. Interestingly, a single dose of the polyanhydride particle-based formulation induced sustained OVA-specific cellular immune responses just as effectively as the prime-boost regimens. In addition, mice receiving single-dose vaccine had similar levels of protection against tumor challenge compared with mice administered prime-boosts. In contrast, measurements of OVA-specific IgG antibody titers indicated that a booster dose was required to stimulate strong humoral immune responses, since it was observed that mice administered a prime-boost vaccine had significantly higher OVA-specific IgG1 serum titers than mice administered a single dose. These findings indicate that the requirement for a booster dose using these particles appears unnecessary for the generation of effective cellular immunity.


Subject(s)
Cancer Vaccines/administration & dosage , Polyanhydrides , Animals , Drug Compounding , Excipients , Female , Immunity, Cellular , Immunity, Humoral , Immunization, Secondary , Immunoglobulin G/analysis , Mice , Mice, Inbred C57BL , Nanoparticles , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/prevention & control , Ovalbumin/administration & dosage , Ovalbumin/immunology , Suspensions
6.
Nanomedicine ; 21: 102055, 2019 10.
Article in English | MEDLINE | ID: mdl-31319179

ABSTRACT

The primary objective of this study was to enhance the antitumor efficacy of a model cancer vaccine through co-delivery of pentaerythritol lipid A (PELA), an immunological adjuvant, and a model tumor antigen, ovalbumin (OVA), separately loaded into polyanhydride particles (PA). In vitro experiments showed that encapsulation of PELA into PA (PA-PELA) significantly enhanced its stimulatory capacity on dendritic cells as evidenced by increased levels of the cell surface costimulatory molecules, CD80/CD86. In vivo experiments showed that PA-PELA, in combination with OVA-loaded PA (PA-OVA), significantly expanded the OVA-specific CD8+ T lymphocyte population compared to PA-OVA alone. Furthermore, OVA-specific serum antibody titers of mice vaccinated with PA-OVA/PA-PELA displayed a significantly stronger shift toward a Th1-biased immune response compared to PA-OVA alone, as evidenced by the substantially higher IgG2C:IgG1 ratios achieved by the former. Analysis of E.G7-OVA tumor growth curves showed that mice vaccinated with PA-OVA/PA-PELA had the slowest average tumor growth rate.


Subject(s)
Cancer Vaccines/pharmacology , Dendritic Cells/drug effects , Immunity, Cellular/drug effects , Neoplasms/drug therapy , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Animals , B7-1 Antigen/immunology , B7-2 Antigen/immunology , CD8-Positive T-Lymphocytes , Cell Proliferation/drug effects , Dendritic Cells/immunology , Humans , Immunity, Cellular/immunology , Immunoglobulin G/immunology , Lipid A/chemistry , Lipid A/pharmacology , Mice , Neoplasms/immunology , Neoplasms/pathology , Polyanhydrides/chemistry , Polyanhydrides/pharmacology , Propylene Glycols/chemistry , Propylene Glycols/pharmacology , Receptors, IgG/immunology , Th1 Cells/drug effects , Th1 Cells/immunology
7.
Mol Pharm ; 11(3): 1022-31, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24494979

ABSTRACT

Chitosan polyplexes containing plasmid DNA (pDNA) have significant potential for pulmonary gene delivery applications. However, prior to using chitosan/pDNA polyplexes (CSpp) in clinical applications, their potential cytotoxicity needs to be investigated. In this study, we formulated 200-400 nm CSpp with amine to phosphate (N/P) ratios that ranged from 1 to 100. We compared two types of plasmids within CSpp: pDNA that was free of CpG sequences (CpG(-)) and pDNA that contained CpG sequences (CpG(+)). Both forms of CSpp showed low cytotoxicity when cultured with A549 and HEK293 cell lines in vitro. CSpp(CpG(-)) generated higher luciferase expression both in vitro, for A549 cells, and in vivo, compared with CSpp(CpG(+)). In addition, CSpp(CpG(-)) elicited milder inflammatory responses in mice one day subsequent to nasal instillation, as determined by proinflammatory cytokine levels within the bronchoalveolar lavage fluid. Our findings suggest that to achieve optimal gene expression with minimal cytotoxicity, inflammation, and oxidative stress, the N/P ratios and CpG sequences in the pDNA of CSpp need to be considered. These findings will inform the preclinical safety assessments of CSpp in pulmonary gene delivery systems.


Subject(s)
Chitosan/chemistry , CpG Islands , DNA/administration & dosage , Gene Transfer Techniques , Plasmids/administration & dosage , Pneumonia/immunology , Transfection/methods , Administration, Intranasal , Animals , Apoptosis , Chitosan/metabolism , Cytokines/metabolism , DNA/chemistry , DNA/metabolism , HEK293 Cells , Humans , Liposomes , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Pneumonia/genetics , Pneumonia/pathology
8.
Int J Pharm ; 638: 122932, 2023 May 10.
Article in English | MEDLINE | ID: mdl-37031810

ABSTRACT

Heterologous prime-boost vaccines have the potential to promote higher immune responses than homologous prime-boost vaccines and were used in this murine study to investigate the effect on the magnitude of the cellular (and humoral) antigen-specific immune responses and antitumor efficacy when a microparticle formulation (prime) is combined with an adenoviral vaccine (boost). Specifically, the prime comprised chick egg ovalbumin (OVA; 25 µg/dose), used here as a model tumor antigen (TA), encapsulated in microparticles (∼700 nm diameter) made from the biodegradable polymer, 50:50 poly(lactic-co-glycolic acid) (PLGA); while attenuated adenovirus (type 5) encoding OVA (Ad5OVA; 108 PFU/dose) was employed as the boost. The ability of OVA-loaded microparticles to enhance OVA-specific antibody responses, OVA-specific CD3 + CD8 + T cell responses and antitumor activity (i.e., protection against OVA-expressing tumor-challenge) to the heterologous prime-boost vaccine was investigated; and it was found that this prime-boost combination could significantly enhance OVA-specific cellular responses compared to all other vaccination groups and was the only group to confer a significant survival advantage over the unvaccinated group (naïve) in a prophylactic animal tumor model. This finding illustrates the potential for combining TA-loaded PLGA-based microparticles with other vaccine formats to improve tumor-specific cellular immune responses.


Subject(s)
Neoplasms , Viral Vaccines , Mice , Animals , Adenoviridae/genetics , Antigens , Immunity, Cellular
9.
Eur J Pharm Biopharm ; 183: 1-12, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36549400

ABSTRACT

Over the last decade, the potential for silver nanoparticles (AgNP) to be used as an anti-melanoma agent has been supported by both in vitro and in vivo evidence. However, an undesirably high concentration of AgNP is often required to achieve an antitumor effect. Therefore a combination treatment that can maintain or improve antitumor efficacy (with lower amounts of AgNP) while also reducing off-target effects is sought. In this study, the combination of AgNP and resiquimod (RSQ: a Toll-like receptor agonist) was investigated and shown to significantly prolong the survival of melanoma-challenged mice when added sequentially. Results from toxicity studies showed that the treatment was non-toxic in mice. Immune cell depletion studies suggested the possible involvement of CD8+ T cells in the antitumor response observed in the AgNP + RSQ (sequential) treatment. NanoString was also employed to further understand the mechanism underlying the increase in the treatment efficacy of AgNP + RSQ (sequential); showing significant changes, compared to the naive group, in gene expression in pathways involved in apoptosis and immune stimulation. In conclusion, the combination of AgNP and RSQ is a new combination worthy of further investigation in the context of melanoma treatment.


Subject(s)
Melanoma , Metal Nanoparticles , Mice , Animals , Silver , CD8-Positive T-Lymphocytes , Melanoma/drug therapy
10.
AAPS J ; 25(4): 57, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37266761

ABSTRACT

Melanoma is the deadliest form of skin cancer and surgery is currently the most effective treatment. However, there are situations where surgery fails or is not an option to treat melanoma patients. Immunotherapy such as immune checkpoint blockade (e.g., anti-PD-1) can be effective as an alternative treatment for melanoma patients; however, the percentage of melanoma patients that exhibit complete responses from anti-PD-1 monotherapy is low, and a hostile immunosuppressive tumor microenvironment may be at least partly responsible. Resiquimod (RSQ) is an imidazoquinolinamine derivative and TLR-7/8 agonist that could enhance the antitumor activity of immune checkpoint blockade when these agents are combined as a treatment for melanoma. Here, the effect of combining systemic anti-PD-1 and locally administered RSQ on the survival of melanoma-challenged mice was tested. Our results demonstrated that anti-PD-1 in combination with RSQ can significantly prolong the survival of melanoma-challenged mice, compared to untreated mice and mice treated with anti-PD-1 alone. In addition, the in vitro studies showed that RSQ can mediate a direct anti-proliferative effect on melanoma cells. In conclusion, the combination of RSQ and anti-PD-1 may be a promising treatment for melanoma patients, especially as both treatments have already been used independently to safely treat melanoma patients.


Subject(s)
Melanoma , Skin Neoplasms , Mice , Animals , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/drug therapy , Melanoma/pathology , Skin Neoplasms/drug therapy , Immunotherapy/methods , Tumor Microenvironment
12.
Pharmaceutics ; 14(10)2022 Sep 29.
Article in English | MEDLINE | ID: mdl-36297510

ABSTRACT

Melanoma is the most lethal form of skin cancer and surgery remains the preferred and most effective treatment. Nevertheless, there are cases where surgery is not a viable method and alternative treatments are therefore adopted. One such treatment that has been tested is topical 5% imiquimod (IMQ) cream, which, although showing promise as a treatment for melanoma, has been found to have undesirable off-target effects. Resiquimod (RSQ) is an immunomodulatory molecule that can activate immune responses by binding to Toll-like receptors (TLR) 7 and 8 and may be more effective than IMQ in the context of melanoma treatment. RSQ can cross the stratum corneum (SC) easily without requiring pretreatment of the skin. In a gel formulation, RSQ has been studied as a monotherapy and adjuvant for melanoma treatment in pre-clinical studies and as an adjuvant in clinical settings. Although side effects of RSQ in gel formulation were also reported, they were never severe enough for the treatment to be suspended. In this review, we discuss the potential use of RSQ as an adjuvant for melanoma treatment.

13.
Methods Mol Biol ; 2455: 319-332, 2022.
Article in English | MEDLINE | ID: mdl-35213004

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is an aggressive liver disease that is considered a major cause of liver cirrhosis and hepatocellular carcinoma. NASH is characterized by multiple underlying genetic mutations, with no approved cure to date. Gene therapies that target those genetic mutations may play a major role in treating this disease, once delivered specifically to the hepatocytes. In this chapter we present, in detail, the synthesis and the characterization of an efficient gene delivery system capable of targeting hepatocytes by exploiting the overexpression of asialoglycoprotein receptors on their cell surface. The targeting ligand, galactose derivative, lactobionic acid (Gal), is first conjugated to bifunctional poly(ethylene glycol) (PEG), and then the formed PEG-Gal is further conjugated to the positively charged polymer, poly(amidoamine) (PAMAM) to form a PAMAM-PEG-Gal construct that can complex and deliver genetic material (e.g., pDNA, siRNA, mRNA) specifically to hepatocytes. We first synthesize PAMAM-PEG-Gal using carbodiimide click chemistry. The synthesized conjugate is characterized using 1H NMR spectroscopy and mass spectrometry. Next, nanoplexes are prepared by combining the positively charged conjugate and the negatively charged genetic material at different nitrogen to phosphate (N/P) ratios; then the size, charge, electrophoretic mobility, and surface morphology of those nanoplexes are estimated. The simplicity of complexing our conjugate with any type of genetic material, the ability of our delivery system to overcome the current limitations of delivering naked genetic material, and the efficiency of delivering its payload specifically to hepatocytes, makes our formulation a promising tool to treat any type of genetic abnormality that arises in hepatocytes, and specifically NASH.


Subject(s)
Gene Transfer Techniques , Liver Neoplasms , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Polyamines/chemistry , Polyethylene Glycols/chemistry
14.
Drug Deliv Transl Res ; 12(7): 1684-1696, 2022 07.
Article in English | MEDLINE | ID: mdl-34635984

ABSTRACT

Endometrial cancer is the most common gynecological cancer that affects the female reproductive organs. The standard therapy for EC for the past two decades has been chemotherapy and/or radiotherapy. PD98059 is a reversible MEK inhibitor that was found in these studies to increase the cytotoxicity of paclitaxel (PTX) against human endometrial cancer cells (Hec50co) in a synergistic and dose-dependent manner. Additionally, while PD98059 arrested Hec50co cells at the G0/G1 phase, and PTX increased accumulation of cells at the G2/M phase, the combination treatment increased accumulation at both the G0/G1 and G2/M phases at low PTX concentrations. We recently developed poly(lactide-co-glycolide) (PLGA) nanoparticles (NPs) modified with polyethylene glycol (PEG) and coated with polyamidoamine (PAMAM) (referred to here as PGM NPs) which have favorable biodistribution profiles in mice, compared to PD98059 solution. Here, in order to enhance tissue distribution of PD98059, PD98059-loaded PGM NPs were prepared and characterized. The average size, zeta potential, and % encapsulation efficiency (%EE) of these NPs was approximately 184 nm, + 18 mV, and 23%, respectively. The PD98059-loaded PGM NPs released ~ 25% of the total load within 3 days in vitro. In vivo murine studies revealed that the pharmacokinetics and biodistribution profile of intravenous (IV) injected PD98059 was improved when delivered as PD98059-loaded PGM NPs as opposed to soluble PD98059. Further investigation of the in vivo efficacy and safety of this formulation is expected to emphasize the potential of its clinical application in combination with commercial PTX formulations against different cancers.


Subject(s)
Endometrial Neoplasms , Nanoparticles , Animals , Cell Line, Tumor , Drug Carriers , Endometrial Neoplasms/drug therapy , Female , Flavonoids , Humans , Mice , Mitogen-Activated Protein Kinase Kinases , Paclitaxel , Polyamines , Polyethylene Glycols , Protein Kinase Inhibitors , Tissue Distribution
15.
Sci Adv ; 8(29): eabk3150, 2022 Jul 22.
Article in English | MEDLINE | ID: mdl-35857851

ABSTRACT

In clinical settings, cancer vaccines as monotherapies have displayed limited success compared to other cancer immunotherapeutic treatments. Nanoscale formulations have the ability to increase the efficacy of cancer vaccines by combatting the immunosuppressive nature of the tumor microenvironment. Here, we have synthesized a previously unexplored cationic polymeric nanoparticle formulation using polyamidoamine dendrimers and poly(d,l-lactic-co-glycolic acid) that demonstrate adjuvant properties in vivo. Tumor-challenged mice vaccinated with an adenovirus-based cancer vaccine [encoding tumor-associated antigen (TAA)] and subsequently treated with this nanoparticulate formulation showed significant increases in TAA-specific T cells in the peripheral blood, reduced tumor burden, protection against tumor rechallenge, and a significant increase in median survival. An investigation into cell-based pathways suggests that administration of the nanoformulation at the site of the developing tumor may have created an inflammatory environment that attracted activated TAA-specific CD8+ T cells to the vicinity of the tumor, thus enhancing the efficacy of the vaccine.

16.
Mol Pharm ; 8(5): 1652-61, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21780831

ABSTRACT

Adjuvants modulate protective CD8(+) T cell responses generated by cancer vaccines. We have previously shown that immunostimulatory cytosine-phosphodiester-guanine (CpG) oligodeoxynucleotide (ODN) significantly augments tumor protection in mice given adenovirus cancer vaccines. Here, we examined the impact of chitosan, another candidate vaccine adjuvant, on protection conferred by adenovirus cancer vaccines. Unexpectedly, immunization of mice with adenovirus cancer vaccines in combination with chitosan provided little protection against tumor challenge. This directly correlated with the reduced detection of Ag-specific CD8(+) T cells, interferon-γ (IFN-γ) production, and cytotoxic T cell activity. We ruled out immunosuppressive regulatory T cells since the frequency did not change regardless of whether chitosan was delivered. In mammalian cell lines, chitosan did not interfere with adenovirus transgene expression. However, infection of primary murine bone marrow-derived dendritic cells with adenovirus complexed with chitosan significantly reduced viability, transgene expression, and upregulation of major histocompatability (MHC) class I and CD86. Our in vitro observations indicate that chitosan dramatically inhibits adenovirus-mediated transgene expression and antigen presenting cell activation, which could prevent CD8(+) T cell activation from occurring in vivo. These surprising data demonstrate for the first time that chitosan vaccine formulations can negatively impact the induction of CD8(+) T cell responses via its effect on dendritic cells, which is clinically important since consideration of chitosan as an adjuvant for vaccine formulations is growing.


Subject(s)
Adenoviridae/immunology , Cancer Vaccines/antagonists & inhibitors , Chitosan/toxicity , Down-Regulation/drug effects , Immunologic Factors/toxicity , T-Lymphocytes, Cytotoxic/drug effects , Adenoviridae/genetics , Animals , Antigen Presentation/drug effects , B7-2 Antigen/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/virology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Genes, Viral/drug effects , Histocompatibility Antigens Class I/metabolism , Interferon-gamma Release Tests , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Transgenes/drug effects
17.
Pharm Res ; 28(2): 215-36, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20721603

ABSTRACT

Therapeutic strategies that involve the manipulation of the host's immune system are gaining momentum in cancer research. Antigen-loaded nanocarriers are capable of being actively taken up by antigen-presenting cells (APCs) and have shown promising potential in cancer immunotherapy by initiating a strong immunostimulatory cascade that results in potent antigen-specific immune responses against the cancer. Such carrier systems offer versatility in that they can simultaneously co-deliver adjuvants with the antigens to enhance APC activation and maturation. Furthermore, modifying the surface properties of these nanocarriers affords active targeting properties to APCs and/or enhanced accumulation in solid tumors. Here, we review some recent advances in these colloidal and particulate nanoscale systems designed for cancer immunotherapy and the potential for these systems to translate into clinical cancer vaccines.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Immunotherapy/methods , Nanomedicine/methods , Nanoparticles/chemistry , Adjuvants, Immunologic/administration & dosage , Animals , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , Drug Carriers , Drug Delivery Systems , Humans , Mice , Neoplasms/immunology , Neoplasms/therapy
18.
AAPS J ; 23(1): 19, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33404992

ABSTRACT

Malignant melanoma is an aggressive form of skin cancer for which there is currently no reliable therapy and is considered one of the leading health issues in the USA. At present, surgery is the most effective and acceptable treatment; however, surgical excision can be impractical in certain circumstances. Topical skin delivery of drugs using topical formulations is a potential alternative approach which can have many advantages aside from being a non-invasive delivery route. Nevertheless, the presence of the stratum corneum (SC) limits the penetration of drugs through the skin, lowering their treatment efficacy and raising concerns among physicians and patients as to their effectiveness. Currently, research groups are trying to circumvent the SC barrier by using skin penetration enhancement (SPE) strategies. The SPE strategies investigated include chemical skin penetration enhancers (CPEs), physical skin penetration enhancers (PPEs), nanocarrier systems, and a combination of SPE strategies (cream). Of these, PPEs and cream are the most advanced approaches in terms of preclinical and clinical studies, respectively.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Epidermis/metabolism , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Administration, Cutaneous , Antineoplastic Agents/pharmacokinetics , Chemistry, Pharmaceutical , Drug Evaluation, Preclinical , Epidermis/pathology , Humans , Melanoma/pathology , Nanoparticles/chemistry , Permeability/drug effects , Skin Absorption/drug effects , Skin Cream/administration & dosage , Skin Cream/pharmacokinetics , Skin Neoplasms/pathology
19.
Eur J Pharm Biopharm ; 159: 21-35, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33338604

ABSTRACT

In the field of drug delivery, the most commonly used treatments have traditionally been systemically delivered using oral or intravenous administration. The problems associated with this type of delivery is that the drug concentration is controlled by first pass metabolism, and therefore may not always remain within the therapeutic window. Implantable drug delivery systems (IDDSs) are an excellent alternative to traditional delivery because they offer the ability to precisely control the drug release, deliver drugs locally to the target tissue, and avoid the toxic side effects often experienced with systemic administration. Since the creation of the first FDA-approved IDDS in 1990, there has been a surge in research devoted to fabricating and testing novel IDDS formulations. The versatility of these systems is evident when looking at the various biomedical applications that utilize IDDSs. This review provides an overview of the history of IDDSs, with examples of the different types of IDDS formulations, as well as looking at current and future biomedical applications for such systems. Though there are still obstacles that need to be overcome, ever-emerging new technologies are making the manufacturing of IDDSs a rewarding therapeutic endeavor with potential for further improvements.


Subject(s)
Delayed-Action Preparations/administration & dosage , Drug Implants/administration & dosage , Drug-Eluting Stents/history , Delayed-Action Preparations/pharmacokinetics , Drug Approval/history , Drug Compounding/methods , Drug Compounding/trends , Drug Implants/history , Drug Implants/pharmacokinetics , History, 20th Century , History, 21st Century , Humans , United States , United States Food and Drug Administration
20.
Adv Ther (Weinh) ; 4(7)2021 Jul.
Article in English | MEDLINE | ID: mdl-34423112

ABSTRACT

Different tetrahydrobenzo[b]thiophene derivatives were explored as new tubulin polymerization destabilizers to arrest tumor cell mitosis. A series of compounds incorporating the tetrahydrobenzo[b]thiophene scaffold were synthesized, and their biological activities were investigated. The cytotoxicity of each of the synthesized compounds was assessed against a range of cell lines. Specifically, the benzyl urea tetrahydrobenzo[b]thiophene derivative, 1-benzyl-3-(3-cyano-4,5,6,7-tetrahydrobenzo[b]thiophen-2-yl)urea (BU17), was identified as the most potent compound with broad-spectrum antitumor activity against several cancer cell lines. The potential mechanism(s) of action were investigated where dose-dependent G2/M accumulation and A549 cell cycle arrest were detected. Additionally, A549 cells treated with BU17 expressed enhanced levels of caspase 3 and 9, indicating the induction of apoptosis. Furthermore, it was found that BU17 inhibits WEE1 kinase and targets tubulin by blocking its polymerization. BU17 was also formulated into PLGA nanoparticles, and it was demonstrated that BU17-loaded nanoparticles could significantly enhance antitumor activity compared to the soluble counterpart.

SELECTION OF CITATIONS
SEARCH DETAIL