Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
J Control Release ; 315: 126-138, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31672625

ABSTRACT

In this study, dual drug-loaded nanoparticles were constructed to co-deliver low-dose doxorubicin (DOX) and miR-200c (DOX/miR-NPs) to inhibit programmed death-1 receptor (PD-L1) expression and trigger immunogenic cell death (ICD) in cancer cells. Two block copolymers, folic acid (FA)-conjugated PLGA-PEG (PLGA-PEG-FA) and PLGA-PEI, were formulated as folate-targeted NPs and loaded with DOX and miR-200c. The NPs, which were formed as nanosize objects (110.4 ± 2.1) with narrow size distribution (0.19 ± 0.02), effectively protected the miR-200c from degradation in serum. Modifying the NPs with FA increased not only their uptake by cancer cells in vitro but also their accumulation in tumor microenvironments in vivo, as compared with those properties of non-FA-modified NPs. The DOX/miR-NPs also exhibited efficacious inhibition of PD-L1 expression and robust induction of ICD in cancer cells in vitro and in vivo, resulting in increased dendritic cell maturation and CD8+ T cell response towards cancer cells. Furthermore, tumor growth was significantly inhibited by folate-targeted NPs loaded with the low-dose DOX/miR-200c combination, but not by treatments with free DOX, miR-NPs or DOX-NPs. Thus, our results suggest that simultaneous PD-L1 inhibition via microRNAs and the induction of an immunogenic tumor microenvironment via low-dose cytotoxic drugs may improve cancer therapy efficacy.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , MicroRNAs/administration & dosage , Nanoparticles , Animals , Antibiotics, Antineoplastic/immunology , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Doxorubicin/immunology , Drug Carriers/chemistry , Drug Delivery Systems , Female , Folic Acid/chemistry , Humans , Immunogenic Cell Death/immunology , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/immunology , Polyesters/chemistry , Polyethylene Glycols/chemistry , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
2.
ACS Appl Mater Interfaces ; 11(10): 9850-9859, 2019 Mar 13.
Article in English | MEDLINE | ID: mdl-30788951

ABSTRACT

Chemo-immunotherapy is an important tool to overcome tumor immune suppression in cancer immunotherapy. Herein, we report a surface-layer (S-layer) protein-enhanced immunotherapy strategy based on cell membrane-coated S-CM-HPAD nanoparticles for the effective malignant tumor therapy and metastasis inhibition. The S-CM-HPAD NPs could effectively deliver the tumor antigen, DOX, and immunoadjuvant to the homotypic tumor by the homotypic targeting ability of the coated cell membrane. In addition to its ability to induce tumor cell death, the loaded DOX could enhance the immunotherapy response by inhibition of myeloid-derived suppressor cells (MDSCs). Because of the intrinsic adjuvant property and capability to surface display epitopes and proteins, the S-layers localized on the surface of S-CM-HPAD NPs potentiated the immune response to the antigen. The results confirmed that the protective immunity against tumor occurrence was promoted effectively by prompting proliferation of lymphocytes and secretion of cytokine caused by the tumor-associated antigen and adjuvant. The excellent combinational therapeutic effects on the inhibition of tumor growth and metastasis in the melanoma tumor models demonstrated that the S-layer-enhanced immunotherapeutic method is a promising strategy for tumor immunotherapy of malignant tumor growth and metastasis.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cell Proliferation/drug effects , Immunoconjugates/administration & dosage , Melanoma, Experimental/drug therapy , Nanoparticles/administration & dosage , Animals , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/chemistry , Cell Membrane/chemistry , Cell Membrane/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Doxorubicin/immunology , Humans , Immunity, Cellular/drug effects , Immunoconjugates/chemistry , Immunoconjugates/immunology , Melanoma, Experimental/pathology , Membrane Proteins/chemistry , Membrane Proteins/immunology , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Nanoparticles/chemistry , Neoplasm Metastasis , Surface Properties
3.
Theranostics ; 8(11): 3164-3175, 2018.
Article in English | MEDLINE | ID: mdl-29896310

ABSTRACT

Rationale: Increasing frequency of human exposure to PEG-related products means that healthy people are likely to have pre-existing anti-PEG antibodies (pre-αPEG Ab). However, the influence of pre-αPEG Abs on the pharmacokinetics (PK) and therapeutic efficacy of LipoDox is unknown. Methods: We generated two pre-αPEG Ab mouse models. First, naïve mice were immunized with PEGylated protein to generate an endogenous αPEG Ab titer (endo αPEG). Second, monoclonal αPEG Abs were passively transferred (αPEG-PT) into naïve mice to establish a αPEG titer. The naïve, endo αPEG and αPEG-PT mice were intravenously injected with 111in-labeled LipoDox to evaluate its PK. Tumor-bearing naïve, endo αPEG and αPEG-PT mice were intravenously injected with 111in-labeled LipoDox to evaluate its biodistribution. The therapeutic efficacy of LipoDox was estimated in the tumor-bearing mice. Results: The areas under the curve (AUC)last of LipoDox in endo αPEG and αPEG-PT mice were 11.5- and 15.6- fold less, respectively, than that of the naïve group. The biodistribution results suggested that pre-αPEG Ab can significantly reduce tumor accumulation and accelerate blood clearance of 111In-labeled LipoDox from the spleen. The tumor volumes of the tumor-bearing endo αPEG and αPEG-PT mice after treatment with LipoDox were significantly increased as compared with that of the tumor-bearing naïve mice. Conclusions: Pre-αPEG Abs were found to dramatically alter the PK and reduce the tumor accumulation and therapeutic efficacy of LipoDox. Pre-αPEG may have potential as a marker to aid development of personalized therapy using LipoDox and achieve optimal therapeutic efficacy.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Antibodies/immunology , Doxorubicin/analogs & derivatives , Neoplasms, Experimental/drug therapy , Animals , Antibiotics, Antineoplastic/immunology , Antibiotics, Antineoplastic/pharmacokinetics , Antibodies/blood , Doxorubicin/immunology , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Female , Liposomes/pharmacokinetics , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/therapeutic use
4.
Bioconjug Chem ; 28(11): 2747-2755, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29090582

ABSTRACT

Opsonization (coating) of nanoparticles with complement C3 component is an important mechanism that triggers immune clearance and downstream anaphylactic and proinflammatory responses. The variability of complement C3 binding to nanoparticles in the general population has not been studied. We examined complement C3 binding to dextran superparamagnetic iron oxide nanoparticles (superparamagnetic iron oxide nanoworms, SPIO NWs, 58 and 110 nm) and clinically approved nanoparticles (carboxymethyl dextran iron oxide ferumoxytol (Feraheme, 28 nm), highly PEGylated liposomal doxorubicin (LipoDox, 88 nm), and minimally PEGylated liposomal irinotecan (Onivyde, 120 nm)) in sera from healthy human individuals. SPIO NWs had the highest variation in C3 binding (n = 47) between subjects, with a 15-30 fold range in levels of C3. LipoDox (n = 12) and Feraheme (n = 18) had the lowest levels of variation between subjects (an approximately 1.5-fold range), whereas Onivyde (n = 18) had intermediate between-subject variation (2-fold range). There was no statistical difference between males and females and no correlation with age. There was a significant correlation in complement response between small and large SPIO NWs, which are similar structurally and chemically, but the correlations between SPIO NWs and other types of nanoparticles, and between LipoDox and Onivyde, were not significant. The calculated average number of C3 molecules bound per nanoparticle correlated with the hydrodynamic diameter but was decreased in LipoDox, likely due to the PEG coating. The conclusions of this study are (1) all nanoparticles show variability of C3 opsonization in the general population; (2) an individual's response toward one nanoparticle cannot be reliably predicted based on another nanoparticle; and (3) the average number of C3 molecules per nanoparticle depends on size and surface coating. These results provide new strategies to improve nanomedicine safety.


Subject(s)
Antibiotics, Antineoplastic/immunology , Complement C3/immunology , Dextrans/immunology , Doxorubicin/analogs & derivatives , Ferrosoferric Oxide/immunology , Liposomes/immunology , Adult , Antineoplastic Agents, Phytogenic/administration & dosage , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Complement Activation , Doxorubicin/immunology , Female , Humans , Irinotecan , Magnetite Nanoparticles , Male , Middle Aged , Polyethylene Glycols , Surface Properties
5.
Anal Chem ; 88(21): 10661-10666, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27726379

ABSTRACT

Polyethylene glycol (PEG) is a biocompatible polymer that is often attached to therapeutic molecules to improve bioavailability and therapeutic efficacy. Although antibodies with specificity for PEG may compromise the safety and effectiveness of PEGylated medicines, the prevalence of pre-existing anti-PEG antibodies in healthy individuals is unclear. Chimeric human anti-PEG antibody standards were created to accurately measure anti-PEG IgM and IgG antibodies by direct ELISA with confirmation by a competition assay in the plasma of 1504 healthy Han Chinese donors residing in Taiwan. Anti-PEG antibodies were detected in 44.3% of healthy donors with a high prevalence of both anti-PEG IgM (27.1%) and anti-PEG IgG (25.7%). Anti-PEG IgM and IgG antibodies were significantly more common in females as compared to males (32.0% vs 22.2% for IgM, p < 0.0001 and 28.3% vs 23.0% for IgG, p = 0.018). The prevalence of anti-PEG IgG antibodies was higher in younger (up to 60% for 20 year olds) as opposed to older (20% for >50 years) male and female donors. Anti-PEG IgG concentrations were negatively associated with donor age in both females (p = 0.0073) and males (p = 0.026). Both anti-PEG IgM and IgG strongly bound PEGylated medicines. The described assay can assist in the elucidation of the impact of anti-PEG antibodies on the safety and therapeutic efficacy of PEGylated medicines.


Subject(s)
Immunoglobulin G/blood , Immunoglobulin M/blood , Polyethylene Glycols/metabolism , Adult , Aged , Aged, 80 and over , Animals , Asian People , Doxorubicin/analogs & derivatives , Doxorubicin/immunology , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Interferon-alpha/immunology , Male , Mice , Middle Aged , Recombinant Fusion Proteins/immunology , Recombinant Proteins/immunology , Young Adult
6.
J Control Release ; 221: 1-8, 2016 Jan 10.
Article in English | MEDLINE | ID: mdl-26608877

ABSTRACT

In order to improve patient's benefit and safety, comprehensive regulatory guidelines on specificities of Non-Biological Complex Drugs (NBCDs), such as doxorubicin-encapsulated liposomes, and their follow-on versions are needed. Here, we compare Doxil® and its European analog Caelyx® with the two follow-on products DOXOrubicin (approved by the US Food and Drug Administration) and SinaDoxosome (produced in Iran) by cryogenic transmission electron microscopy, dynamic light scattering and Nanoparticle Tracking Analysis, and assess their potential in activating the complement system in human sera. We found subtle physicochemical differences between the tested liposomal products and even between the tested batches of Doxil® and Caelyx®. Notably, these included differences in vesicular population aspect ratios and particle number. Among the tested products, only SinaDoxosome, in addition to the presence of unilamellar vesicles with entrapped doxorubicin crystals, contained empty circular disks. Differences were also found in complement responses, which may be related to some morphological differences. This study has demonstrated an integrated biophysical and immunological toolbox for improved analysis and detection of physical differences among vesicular populations that may modulate their clinical performance. Combined, these approaches may help better product selection for infusion to the patients as well as for improved design and characterization of future vesicular NBCDs with enhanced clinical performance and safety.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/immunology , Complement Activation/drug effects , Doxorubicin/analogs & derivatives , Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Doxorubicin/immunology , Humans , Liposomes/chemistry , Liposomes/immunology , Liposomes/ultrastructure , Particle Size , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry
7.
Biol Pharm Bull ; 38(3): 417-24, 2015.
Article in English | MEDLINE | ID: mdl-25757923

ABSTRACT

PEGylation, which is the surface modification of nanocarriers with polyethylene glycol (PEG), has increased the circulation time and reduced the immunogenic responses to nanocarriers. However, many reports have demonstrated that the intravenous injection of sterically stabilized PEGylated liposome (SL) causes an accelerated blood clearance (ABC) of subsequent doses via anti-PEG immunoglobulin M (IgM)-mediated complement activation. In the present study, the relationships between serum anti-PEG IgM concentration, the intensity of complement activation and the hepatic clearance of SL were quantitatively investigated for their role in the ABC phenomenon. Interestingly, with increasing serum anti-PEG IgM concentrations, the intensity of complement activation increased linearly, while the intensity of the hepatic clearance of SL was increased and then saturated. In addition, only 15-17% of anti-PEG IgM in blood circulation induced by SL at different doses was associated with a second dose SL. The present results indicate that it is the hepatic uptake of SL that is the limiting step in the ABC phenomenon, rather than the association of anti-PEG IgM to the SL and a subsequent complement activation.


Subject(s)
Complement System Proteins/metabolism , Doxorubicin/analogs & derivatives , Drug Carriers , Immunoglobulin M/blood , Liver/metabolism , Polyethylene Glycols/metabolism , Animals , Doxorubicin/administration & dosage , Doxorubicin/immunology , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Carriers/adverse effects , Injections, Intravenous , Liposomes , Male , Mice, Inbred BALB C , Nanoparticles , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/pharmacology
8.
Salvador; s.n; 2015. 96 p. ilus.
Thesis in Portuguese | LILACS | ID: biblio-870338

ABSTRACT

Introdução: A doxorrubicina (DOX) é um quimioterápico antracíclico amplamente usado para o tratamento de diversos tumores humanos, entretanto, o desenvolvimento de reações adversas à droga, em particular, cardiotoxicidade, tem limitado seu uso. Embora a toxicidade cardíaca induzida pela DOX pareça ser multifatorial, a hipótese mais investigada tem sido a formação de espécies reativas de oxigênio (ROS) e há evidências apontando para as mitocôndrias cardíacas como alvos primários da toxicidade da DOX. Esse dano oxidativo pode iniciar peroxidação lipídica e pode ser potencialmente limitado pelo uso de antioxidantes. Objetivo: O objetivo do presente estudo foi avaliar a possível eficácia do ácido lipoico (AL) e do Mito-TEMPO (Mito-T) como agentes protetores contra a cardiotoxicidade induzida pela DOX in vitro e in vivo e investigar se essa proteção pode afetar a atividade antitumoral da DOX. Método e Resultados: A capacidade do AL e Mito-T eliminar radicais livres foi avaliada usando o teste do 2,2-difenil-1-picril-hidrazila (DPPH). Menor atividade antioxidante do AL (29%) comparada ao Mito-T (63%) foi observada. DOX reduziu a viabilidade de células H9c2 (CI50 = 40,83 M, IC 95% = 28,64 – 58,21 M) e aumentou a concentração de malondialdeído (MLDA), um marcador de peroxidação lipídica, confirmando a citotoxicidade induzida pela DOX in vitro. O pré-tratamento com AL ou Mito-T não promoveu proteção contra o dano induzido pela DOX in vitro. Uma única injeção intraperitoneal (i.p.) de DOX (24 mg/kg de peso corpóreo) induziu redução significante no peso corpóreo (p<0,001), elevação da atividade sérica total de creatina quinase (p<0,05) e creatina quinase-MB (p<0,05), aumento na concentração de malondialdeído em mitocôndrias (p<0,05) e tecido cardíaco (p<0,01) em camundongos da linhagem C57BL/6 após 48 horas. O pré-tratamento dos animais com Mito-T (5 mg/kg de peso corpóreo, i.p., por dois dias, 48 e 24 horas antes da DOX) reduziu significativamente a peroxidação lipídica de mitocôndrias cardíacas (p<0,01) indicando o direcionamento do antioxidante para a mitocôndria. O tratamento com Mito-T ou AL, duas vezes, 24 e uma hora antes do tratamento com DOX, inibiu a atividade sérica de creatina quinase total (p<0,05). Além disso, o tratamento de camundongos apresentando tumor B16F10 com AL não interferiu na eficácia antitumoral da DOX. Conclusão: Os dados sugerem que a combinação de AL com DOX pode ser benéfica para o tratamento de câncer, entretanto, são necessárias novas investigações para confirmar essa suposição.


Introduction: Doxorubicin (DOX) is an anthracycline chemotherapeutic that is widely used for the treatment of many human tumours, however, the development of adverse drug reactions in particular cardiotoxicity has limited its use. Although doxorubicin-induced cardiac toxicity appears to be multifactorial, the most thoroughly investigated hypothesis has been the formation of reactive oxygen species (ROS) and there is evidence pointing to cardiac mitochondria as primary targets of the toxicity of DOX. This oxidative injury can initiate lipidic peroxidation and may be potentially limited by the use of antioxidants. Aim: The aim of the present study was to evaluate the possible efficacy of lipoic acid (LA) and Mito-TEMPO (Mito-T) as a protective agent against DOX-induced cardiotoxicity in vitro and in vivo and to investigate whether this protection may affect the antitumor activity of DOX. Method and Results: Free radical scavenging capacity of LA and Mito-T was assayed using 1,1-diphenyl-2-picrylhydrazy (DPPH) assay. Lower antioxidant activity for LA (29%) compared to Mito-T (63%) were observed. DOX reduced H9c2 viability (IC50 = 40.83 M, 95% CI = 28.64 – 58.21 M) and increased the levels of malondialdehyde (MLDA), a marker of lipid peroxidation, confirming DOX-induced cytotoxicity in vitro. Pretreatment with LA or Mito-T did not provide protection against DOX-induced damage in vitro. A single intraperitoneal (i.p.) injection of DOX (24 mg/kg body weight) induced a significant reduction in body weight (p<0.001), elevation of serum activity of total creatine kinase (p<0.05) and creatine kinase-MB (p<0.05), increase in malondialdehyde levels in cardiac mitochondria (p<0.05) and cardiac tissue (p<0.01) in C57BL/6 mice after 48 hours...


Subject(s)
Humans , Thioctic Acid/administration & dosage , Thioctic Acid/analysis , Thioctic Acid/immunology , Thioctic Acid/therapeutic use , Doxorubicin/immunology , Doxorubicin/supply & distribution , Doxorubicin/toxicity , Doxorubicin/therapeutic use
9.
Drug Discov Today ; 19(12): 1945-52, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25205349

ABSTRACT

Conjugation of polyethylene glycol (PEG) to therapeutics has proven to be an effective approach to increase the serum half-life. However, the increased use of PEGylated therapeutics has resulted in unexpected immune-mediated side-effects. There are claims that these are caused by anti-PEG antibodies inducing rapid clearance. These claims are however hampered by the lack of standardized and well-validated antibody assays. PEGylation has also been associated with the activation of the complement system causing severe hypersensitivity reactions. Here, we critically review the clinical and analytical tools used. In addition, we propose an explanation of the immune-mediated side-effects of PEGylated products based on the haptogenic properties of PEG, responsible for complement activation and the induction of anti-PEG antibodies.


Subject(s)
Antibodies/immunology , Asparaginase/immunology , Doxorubicin/analogs & derivatives , Peptides/immunology , Urate Oxidase/immunology , Animals , Asparaginase/therapeutic use , Complement Activation , Doxorubicin/immunology , Doxorubicin/therapeutic use , Humans , Interferon-alpha/immunology , Interferon-alpha/therapeutic use , Peptides/therapeutic use , Polyethylene Glycols/therapeutic use , Urate Oxidase/therapeutic use
10.
Gynecol Oncol ; 133(3): 584-90, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24680909

ABSTRACT

OBJECTIVE: Women with BRCA-associated ovarian cancer demonstrate excellent responses to Pegylated Liposomal Doxorubicin (PLD). PLD has also been shown to enhance T cell recognition of tumor cells. Here we characterize immunophenotypic changes associated with BRCA1 dysfunction in ovarian cancer cells, and evaluate the T cell contribution to the therapeutic efficacy of PLD in a BRCA1- ovarian cancer model to determine whether enhanced anti-tumor immunity contributes to the improved response to PLD in BRCA1- ovarian cancers. METHODS: The immunophenotype of BRCA1- and wild-type (WT) ovarian cancer cells and their response to PLD were compared in vitro using flow cytometry. T cell recruitment to BRCA1- tumors was evaluated with flow cytometry and immunohistochemistry. The contribution of T cell populations to the therapeutic effect of PLD in a BRCA1- model was evaluated using immunodepleting antibodies with PLD in vivo. RESULTS: The cytotoxic response to PLD was similar in BRCA1- and WT cells in vitro. BRCA1- inactivation resulted in higher expression of Fas and MHC-I at baseline and after PLD exposure. PLD prolonged the survival of BRCA1- tumor bearing mice and increased intratumoral T cell recruitment. CD4+ depletion combined with PLD significantly prolonged overall survival (p=0.0204) in BRCA1- tumor-bearing mice. CONCLUSION: Differences in the immunophenotype of BRCA1- and WT cells are amplified by PLD exposure. The enhanced immunomodulatory effects of PLD in BRCA1- tumors may be exploited therapeutically by eliminating suppressive CD4+ T cells. Our results support further study of combination therapy using PLD and immune agents, particularly in women with BRCA gene mutations.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/analogs & derivatives , Genes, BRCA1 , Immunomodulation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Ovarian Neoplasms/genetics , T-Lymphocytes/drug effects , Animals , Antibiotics, Antineoplastic/immunology , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Doxorubicin/immunology , Doxorubicin/pharmacology , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Ovarian Neoplasms/immunology , Polyethylene Glycols/pharmacology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
11.
Proc Natl Acad Sci U S A ; 110(43): 17253-8, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24101477

ABSTRACT

Phenotypic polarization of macrophages is regulated by a milieu of cues in the local tissue microenvironment. Although much is known about how soluble factors influence macrophage polarization, relatively little is known about how physical cues present in the extracellular environment might modulate proinflammatory (M1) vs. prohealing (M2) activation. Specifically, the role of cell shape has not been explored, even though it has been observed that macrophages adopt different geometries in vivo. We and others observed that macrophages polarized toward different phenotypes in vitro exhibit dramatic changes in cell shape: M2 cells exhibit an elongated shape compared with M1 cells. Using a micropatterning approach to control macrophage cell shape directly, we demonstrate here that elongation itself, without exogenous cytokines, leads to the expression of M2 phenotype markers and reduces the secretion of inflammatory cytokines. Moreover, elongation enhances the effects of M2-inducing cytokines IL-4 and IL-13 and protects cells from M1-inducing stimuli LPS and IFN-γ. In addition shape- but not cytokine-induced polarization is abrogated when actin and actin/myosin contractility are inhibited by pharmacological agents, suggesting a role for the cytoskeleton in the control of macrophage polarization by cell geometry. Our studies demonstrate that alterations in cell shape associated with changes in ECM architecture may provide integral cues to modulate macrophage phenotype polarization.


Subject(s)
Biomarkers/analysis , Cell Shape/immunology , Cytokines/immunology , Inflammation Mediators/immunology , Macrophages/immunology , Amides/pharmacology , Animals , Arginase/immunology , Arginase/metabolism , Azepines/pharmacology , Biomarkers/metabolism , Cell Polarity/drug effects , Cell Polarity/immunology , Cell Shape/drug effects , Cells, Cultured , Cytochalasin D/pharmacology , Cytokines/metabolism , Doxorubicin/analogs & derivatives , Doxorubicin/immunology , Doxorubicin/metabolism , Female , Flow Cytometry , Immunophenotyping/methods , Inflammation Mediators/metabolism , Interferon-gamma/pharmacology , Interleukin-13/pharmacology , Interleukin-4/pharmacology , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/metabolism , Mannose Receptor , Mannose-Binding Lectins/immunology , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/metabolism , Pyridines/pharmacology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism
12.
J Control Release ; 160(2): 382-7, 2012 Jun 10.
Article in English | MEDLINE | ID: mdl-22421426

ABSTRACT

PEGylated liposomal doxorubicin (Doxil) has been used in cancer chemotherapy for 16 years. Clinical experience shows that it can cause mild-to-severe hypersensitivity (infusion) reactions, which are manifestations of complement (C) activation-related pseudoallergy (CARPA). Although in most cases CARPA is inconsequential, a main symptom, cardiopulmonary distress, may be life threatening in hypersensitive individuals. To date, the prevention of Doxil-induced CARPA is based on premedication and a slow infusion protocol. The present study suggests desensitization by Doxil-like empty liposomes, called placebo Doxil (Doxebo), as an alternative strategy, which is based on the tachyphylactic nature of Doxil reactions. Doxebo-induced tolerance to Doxil was shown to develop within minutes and to be specific to Doxil-like PEGylated liposomes. The procedure of desensitization involves slow, low-dose pre-infusion of Doxebo before Doxil treatment which minimizes the ensuing physiological changes or keeps them subclinical. Although the mechanism of tolerance induction is not yet clear, the effector arm of C response is unlikely to be affected, as the vascular reactivity of desensitized pigs to zymosan remains intact. Desensitization with empty vesicles represents a novel approach for reducing the risk of anaphylactic reactions to drug carrier liposomes. The underlying immediate, most likely passive silencing of an innate immune response may represent a novel mechanism of tolerance induction which may work for other reactogenic nanosystems as well.


Subject(s)
Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Infusions, Intravenous/adverse effects , Placebos/administration & dosage , Polyethylene Glycols/chemistry , Tachyphylaxis/immunology , Animals , Complement Activation/drug effects , Complement Activation/immunology , Desensitization, Immunologic/methods , Doxorubicin/immunology , Drug Carriers/chemistry , Female , Hypersensitivity/etiology , Hypersensitivity/immunology , Hypersensitivity/prevention & control , Infusions, Intravenous/methods , Liposomes , Male , Placebos/chemistry , Swine
13.
Cancer Res ; 71(14): 4809-20, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21646474

ABSTRACT

We show, in a series of established experimental breast adenocarcinomas and fibrosarcomas induced by carcinogen de novo in mice, that the therapeutic efficacy of doxorubicin treatment is dependent on CD8 T cells and IFN-γ production. Doxorubicin treatment enhances tumor antigen-specific proliferation of CD8 T cells in tumor-draining lymph nodes and promotes tumor infiltration of activated, IFN-γ-producing CD8 T cells. Optimal doxorubicin treatment outcome also requires both interleukin (IL)-1ß and IL-17 cytokines, as blockade of IL-1ß/IL-1R or IL-17A/IL-17Rα signaling abrogated the therapeutic effect. IL-23p19 had no observed role. The presence of γδ T cells, but not Jα18(+) natural killer T cells, at the time of doxorubicin treatment was also important. In tumor samples taken from breast cancer patients prior to treatment with anthracycline chemotherapy, a correlation between CD8α, CD8ß, and IFN-γ gene expression levels and clinical response was observed, supporting their role in the therapeutic efficacy of anthracyclines in humans. Overall, these data strongly support the pivotal contribution of both innate and adaptive immunity in treatment outcomes of anthracycline chemotherapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Fibrosarcoma/drug therapy , Fibrosarcoma/immunology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Adaptive Immunity/drug effects , Adaptive Immunity/immunology , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Animals , Antibiotics, Antineoplastic/immunology , CD8 Antigens/biosynthesis , CD8 Antigens/genetics , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Doxorubicin/immunology , Epitopes, T-Lymphocyte/immunology , Female , Fibrosarcoma/genetics , Gene Expression , Immunity, Innate/drug effects , Immunity, Innate/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
14.
J Control Release ; 146(2): 175-81, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20388529

ABSTRACT

Since their introduction, poly(ethylene glycol)-phospholipid (PEG-PL) conjugates have found many applications in design and engineering of nanosized delivery systems for controlled delivery of pharmaceuticals especially to non-macrophage targets. However, there are reports of idiosyncratic reactions to certain PEG-PL engineered nanomedicines in both experimental animals and man. These reactions are classified as pseudoallergy and may be associated with cardiopulmonary disturbance and other related symptoms of anaphylaxis. Recent studies suggest that complement activation may be a contributing, but not a rate limiting factor, in eliciting hypersensitivity reactions to such nanomedicines in sensitive individuals. This is rather surprising since PEGylated structures are generally assumed to suppress protein adsorption and blood opsonization events including complement. Here, we examine the molecular basis of complement activation by PEG-PL engineered nanomedicines and carbon nanotubes and discuss the challenges ahead.


Subject(s)
Anaphylaxis/chemically induced , Complement Activation , Nanostructures/administration & dosage , Nanotubes, Carbon/adverse effects , Phospholipids/metabolism , Polyethylene Glycols/metabolism , Animals , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/immunology , Doxorubicin/adverse effects , Doxorubicin/immunology , Humans , Nanostructures/adverse effects , Phospholipids/adverse effects , Polyethylene Glycols/adverse effects , Serum/drug effects , Serum/immunology , Swine
15.
J Transl Med ; 7: 104, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-20003308

ABSTRACT

BACKGROUND: Liposomal doxorubicin (Doxil) is a cytotoxic chemotherapy drug with a favorable hematologic toxicity profile. Its active drug, doxorubicin, has interesting immunomodulatory properties. Here, the effects of Doxil on surviving tumor cell immunophenotype were investigated. METHODS: Using ID8 murine ovarian cancer cells, the immunomodulatory effects of Doxil were studied by measuring its impact on ovarian cancer cell expression of MHC class-I and Fas, and susceptibility to immune attack in vitro. To evaluate the ability of Doxil to cooperate with cancer immunotherapy, the interaction between Doxil and Interleukin 18 (IL-18), a pleiotropic immunostimulatory cytokine, was investigated in vivo in mice bearing ID8-Vegf tumors. RESULTS: While Doxil killed ID8 tumor cells in a dose-dependent manner, tumor cells escaping Doxil-induced apoptosis upregulated surface expression of MHC-I and Fas, and were sensitized to CTL killing and Fas-mediated death in vitro. We therefore tested the hypothesis that the combination of immunotherapy with Doxil provides positive interactions. Combination IL-18 and Doxil significantly suppressed tumor growth compared with either monotherapy in vivo and uniquely resulted in complete tumor regression and long term antitumor protection in a significant proportion of mice. CONCLUSION: These data demonstrate that Doxil favorably changes the immunophenotype of a large fraction of the tumor that escapes direct killing thus creating an opportunity to expand tumor killing by immunotherapy, which can be capitalized through addition of IL-18 in vivo.


Subject(s)
Antibiotics, Antineoplastic , Doxorubicin , Interleukin-18 , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/immunology , Animals , Antibiotics, Antineoplastic/immunology , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Doxorubicin/immunology , Doxorubicin/therapeutic use , Female , Humans , Immunophenotyping , Interleukin-18/immunology , Interleukin-18/therapeutic use , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Survival Rate
16.
Cancer Immunol Immunother ; 58(1): 49-59, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18446337

ABSTRACT

Abnormal accumulation of myeloid-derived suppressor cells (MDSC) is an important mechanism of tumor immune evasion. Cyclophosphamide (CTX) has also been shown in non-tumor bearing animals to cause transient surges in MDSC. Knowledge of MDSC is primarily based on preclinical work, and to date only few published studies have involved cancer patients. The goal of this study was to test the hypothesis that circulating MDSC levels correlate with clinical cancer stage, CTX-based chemotherapy, and metastatic tumor burden. Whole blood was collected from 106 newly diagnosed solid tumor patients (stages I-IV). Percentages of circulating MDSC (Lin(-/Lo), HLA DR-, CD33(+)CD11b(+)) were determined prior to initiation of systemic therapy. In 17 early stage breast cancer patients receiving doxorubicin-cyclophosphamide chemotherapy every 14 days (ddAC) blood was collected on day 1 of each cycle. Circulating MDSC were significantly increased in cancer patients of all stages relative to healthy volunteers. A significant correlation between circulating MDSC and clinical cancer stage was also observed. Moreover, among stage IV patients, those with extensive metastatic tumor burden had the highest percent and absolute number of MDSC. Significant increases in circulating MDSC were observed with ddAC when compared with pretreatment levels. Circulating MDSC levels correlate with clinical cancer stage, ddAC, and metastatic tumor burden. This information must be incorporated into the design of future trials exploring immune-based therapeutic strategies. Pharmacologic modulation of MDSC should also be tested in future clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/immunology , Breast Neoplasms/immunology , Cyclophosphamide/immunology , Doxorubicin/immunology , Myeloid Cells/cytology , Myeloid Cells/immunology , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/physiopathology , Cyclophosphamide/therapeutic use , Disease Progression , Doxorubicin/therapeutic use , Female , Flow Cytometry , Humans , Male , Middle Aged , Neoplasm Staging , Reference Standards , Tumor Burden/immunology
17.
Nan Fang Yi Ke Da Xue Xue Bao ; 28(8): 1503-5, 2008 Aug.
Article in Chinese | MEDLINE | ID: mdl-18753092

ABSTRACT

OBJECTIVE: To prepare nanospheres coupled with the anti-human liver cancer monoclonal antibody HAb18 and evaluate its immunoreactivity and antitumor effects. METHODS: The nanosphere coupled with the antibody was prepared by intermolecular cross-linking the anti-human liver cancer monoclonal antibody, HAb18, with human serum albumin nanospheres containing ADM [termed HAS(ADM)-NS] via a new hetero-bifunctional cross-linker SPDP. Condensation test and immunofluorescence assay were used to evaluate the immunoreactivity of the nanospheres, and specific binding of HAb18-HAS(ADM)-NS with liver cancer cell line SMMC-7721 was observed with optical and electron microscopes. The specific cytotoxic effects on the target cells were evaluated in vitro by MTT assay. HAb18-HAS(ADM)-NS, HAS(ADM)-NS and ADM were injected separately into nude mice bearing human liver carcinoma to evaluate the inhibitory activity of HAb18-HAS(ADM)-NS in vivo. RESULTS: The immunoreactivity of HAb18-HAS(ADM)-NS was well preserved. HAb18-HAS(ADM)-NS could bind specifically with the SMMC-7721 cells. The IC(50) of HAb18-HAS(ADM)-NS against SMMC-7721 cells was 44.6 microg/ml, lower than that of HAS(ADM)-NS (345.5 microg/ml) and ADM (365.5 microg/ml). The inhibition rate of HAb18-HAS(ADM)-NS on the growth of liver cancer xenografts was significantly higher than that of HAS(ADM)-NS and ADM (P<0.001). CONCLUSION: HAb18-HAS(ADM)-NS has immunoreactivity and can actively and specifically target the liver cancer cells. The antitumor activity of HAb18-HAS(ADM)-NS is significantly higher than that of HAS(ADM)-NS and ADM.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Liver Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/immunology , Cell Line, Tumor , Doxorubicin/administration & dosage , Doxorubicin/immunology , Female , Humans , Immunotoxins/administration & dosage , Immunotoxins/immunology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Nanospheres/administration & dosage , Treatment Outcome , Xenograft Model Antitumor Assays/methods
18.
J Control Release ; 115(3): 251-8, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-17045355

ABSTRACT

The "accelerated blood clearance (ABC) phenomenon", causing PEGylated liposomes to be cleared very rapidly from the circulation upon repeated injection, has been reported to occur in rodents and rhesus monkeys. This rapid clearance was reported to be caused by the binding of PEG-specific IgM, which was generated by the first dose of injected liposomes, to the second dose of liposomes and the subsequent activation of complement, serving in turn as an opsonin. Although there are several PEGylated liposomal formulations, such as Doxil/Caelyx loaded with doxorubicin (DXR), in clinical use, the rapid clearance phenomenon has never been reported for such formulations. In the present article, we report that a first injection of PEGylated liposomes containing encapsulated DXR failed to induce the ABC phenomenon. Likewise, no rapid clearance of the test dose was observed when the first dose of "empty" PEGylated liposomes (without DXR) exceeded 5 micromol phospholipids/kg. By contrast, "empty" PEGylated liposomes at a low dose (1 micromol phospholipids/kg) induced the phenomenon as before. Western blot analysis revealed abundant binding of IgM to PEGylated liposomes when these were incubated in serum from rats that had received "empty" PEGylated liposomes. Substantially less binding of IgM was found when the liposomes were incubated in serum from rats treated with DXR-loaded PEGylated liposomes. For both the empty and the DXR-containing liposomes the amounts of IgM binding to the liposomes decreased with an increasing dose of injected liposomes. Serum obtained from rats following injection of empty PEGylated liposomes caused complement activation by addition of PEGylated liposomes in an inversely dose-dependent manner: the lower the dose, the higher the complement activation. By contrast, no complement activation was detected with serum from rats that had been treated with DXR-loaded PEGylated liposomes. These findings suggest that encapsulation of DXR as well as a relatively high lipid dose abrogate the immune response against PEGylated liposomes which is observed with the same liposomes but without DXR and at low doses. Our observations may thus have important implications for the development, evaluation and therapeutic use of liposomal cytotoxic drug formulations requiring multiple injection schemes.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Liposomes , Polyethylene Glycols/pharmacokinetics , Animals , Antibiotics, Antineoplastic/immunology , Antigen-Antibody Reactions , Chemistry, Pharmaceutical , Cholesterol/administration & dosage , Cholesterol/analogs & derivatives , Cholesterol/pharmacokinetics , Complement Activation , Dose-Response Relationship, Immunologic , Doxorubicin/immunology , Drug Compounding , Immunoglobulin M/blood , Injections, Intravenous , Male , Metabolic Clearance Rate , Phosphatidylethanolamines/pharmacokinetics , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Wistar , Spleen/immunology , Spleen/metabolism , Tissue Distribution , Tritium
19.
Mol Cancer Ther ; 5(9): 2348-57, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16985069

ABSTRACT

Conventional anticancer agents may display antiangiogenic effects, but the underlying mechanism is poorly understood. We determined the antiangiogenic properties of cisplatin, doxorubicin, and the microtubule-targeting agents docetaxel, epothilone B, and vinblastine at concentrations not affecting cell proliferation. We also assessed tubulin and actin morphology and the activity of two key molecules in cell motility, the small Rho GTPases Cdc42 and Rac1. The highest non-toxic concentration (HNTC) of each drug was defined as the concentration inhibiting a maximum of 10% human umbilical vein endothelial cell growth on a 1-hour drug exposure, being for cisplatin 10 micromol/L, doxorubicin 100 nmol/L, docetaxel 10 nmol/L, epothilone B 1 nmol/L, and vinblastine 10 nmol/L. Comparative endothelial cell functional assays using HNTCs for an exposure time of 1 hour indicated that endothelial cell migration in the wound assay, endothelial cell invasion in a transwell invasion system, and endothelial cell formation into tubelike structures on a layer of Matrigel were significantly inhibited by docetaxel, epothilone B, and vinblastine (P < 0.05), but not by cisplatin and doxorubicin. Docetaxel was slightly more efficient in the inhibition of endothelial cell motility than epothilone B and vinblastine. Fluorescence microscopy revealed that only the microtubule-targeting agents affected the integrity of the tubulin and F-actin cytoskeleton, which showed disturbed microtubule structures, less F-actin stress fiber formation, and appearance of nuclear F-actin rings. These observations were associated with early inhibition of Rac1 and Cdc42 activity. In conclusion, HNTCs of microtubule-targeting agents efficiently reduce endothelial cell motility by interference with microtubule dynamics preventing the activation of Rac1/Cdc42 and disorganizing the actin cytoskeleton.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cytoskeleton/drug effects , Endothelial Cells/drug effects , Microtubules/drug effects , Tubulin Modulators/pharmacology , cdc42 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/antagonists & inhibitors , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/physiology , Animals , Cisplatin/pharmacology , Cytoskeleton/metabolism , Doxorubicin/immunology , Endothelial Cells/metabolism , Epothilones/pharmacology , Humans , Mice , Neovascularization, Physiologic/drug effects , Pseudopodia/metabolism , Pseudopodia/physiology , Stress Fibers/metabolism , Stress Fibers/physiology , Vinblastine/pharmacology , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
20.
Cancer Immunol Immunother ; 55(2): 140-50, 2006 Feb.
Article in English | MEDLINE | ID: mdl-15856228

ABSTRACT

We generated a human/mouse chimeric antibody c-SN6j of human IgG1 isotype from a murine anti-human endoglin (EDG) monoclonal antibody (mAb) SN6j that suppressed angiogenesis, tumor growth and metastasis in mice. We determined pharmacokinetics (PKs) and immunogenicity of c-SN6j in monkeys after multiple i.v. injections. A dose-escalation study was performed by administration of c-SN6j into six monkeys at the dose of 1 mg, 3 mg and 10 mg per kg body weight. In addition, both c-SN6j (3 mg/kg) and doxorubicin (0.275 mg/kg) were injected into two monkeys. c-SN6j and doxorubicin were injected twice a week for 3 weeks. We developed a unique and sensitive ELISA by sequentially targeting the common and idiotypic epitopes of c-SN6j-Fv to quantify plasma c-SN6j. Application of the ELISA showed that increasing the c-SN6j dose resulted in a proportional increase in the circulating c-SN6j after the first injection. In addition, the estimated area under the curve (AUC) for the first injection of c-SN6j is proportional to dose. We carried out detailed analyses of PKs of c-SN6j during and after the repeated injections. Our model of PKs fitted the empirical data well. Addition of doxorubicin modulated the PK parameters. We developed two ELISAs to separately determine the immune responses to the murine part and the human part of c-SN6j in monkeys. Interestingly, the murine part induced a weaker immune response than the human part. Doxorubicin potentiated the immune responses. Increasing the dose of c-SN6j increased plasma levels of c-SN6j but did not increase the immune responses to c-SN6j.


Subject(s)
Angiogenesis Inhibitors/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Doxorubicin/pharmacology , Recombinant Fusion Proteins/immunology , Vascular Cell Adhesion Molecule-1/immunology , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacokinetics , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, CD , Dose-Response Relationship, Immunologic , Doxorubicin/administration & dosage , Doxorubicin/immunology , Drug Evaluation, Preclinical , Endoglin , Enzyme-Linked Immunosorbent Assay , Female , Humans , Injections, Intravenous , Macaca fascicularis , Models, Animal , Receptors, Cell Surface , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL