Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 12(6)2020 Jun 08.
Article in English | MEDLINE | ID: mdl-32521631

ABSTRACT

Metastatic breast cancer (MBC) is the leading cause of cancer death in women due to recurrence and resistance to conventional therapies. Thus, MBC represents an important unmet clinical need for new treatments. In this paper we generated a virus-like particle (VLP)-based vaccine (AX09) to inhibit de novo metastasis formation and ultimately prolong the survival of patients with MBC. To this aim, we engineered the bacteriophage MS2 VLP to display an extracellular loop of xCT, a promising therapeutic target involved in tumor progression and metastasis formation. Elevated levels of this protein are observed in a high percentage of invasive mammary ductal tumors including triple negative breast cancer (TNBC) and correlate with poor overall survival. Moreover, xCT expression is restricted to only a few normal cell types. Here, we tested AX09 in several MBC mouse models and showed that it was well-tolerated and elicited a strong antibody response against xCT. This antibody-based response resulted in the inhibition of xCT's function in vitro and reduced metastasis formation in vivo. Thus, AX09 represents a promising novel approach for MBC, and it is currently advancing to clinical development.

2.
Viruses ; 12(5)2020 04 27.
Article in English | MEDLINE | ID: mdl-32349216

ABSTRACT

Virus-like particles (VLP) spontaneously assemble from viral structural proteins. They are naturally biocompatible and non-infectious. VLP can serve as a platform for many potential vaccine epitopes, display them in a dense repeating array, and elicit antibodies against non-immunogenic substances, including tumor-associated self-antigens. Genetic or chemical conjugation facilitates the multivalent display of a homologous or heterologous epitope. Most VLP range in diameter from 25 to 100 nm and, in most cases, drain freely into the lymphatic vessels and induce antibodies with high titers and affinity without the need for additional adjuvants. VLP administration can be performed using different strategies, regimens, and doses to improve the immunogenicity of the antigen they expose on their surface. This article summarizes the features of VLP and presents them as a relevant platform technology to address not only infectious diseases but also chronic diseases and cancer.


Subject(s)
Cancer Vaccines/immunology , Neoplasms/immunology , Vaccines, Virus-Like Particle/immunology , Animals , Antibodies/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Humans , Neoplasms/genetics , Neoplasms/prevention & control , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics
3.
Cancer Immunol Immunother ; 68(1): 131-141, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29947961

ABSTRACT

Tumor relapse and metastatic spreading act as major hindrances to achieve complete cure of breast cancer. Evidence suggests that cancer stem cells (CSC) would function as a reservoir for the local and distant recurrence of the disease, due to their resistance to radio- and chemotherapy and their ability to regenerate the tumor. Therefore, the identification of appropriate molecular targets expressed by CSC may be critical in the development of more effective therapies. Our studies focused on the identification of mammary CSC antigens and on the development of CSC-targeting vaccines. We compared the transcriptional profile of CSC-enriched tumorspheres from an Her2+ breast cancer cell line with that of the more differentiated parental cells. Among the molecules strongly upregulated in tumorspheres we selected the transmembrane amino-acid antiporter xCT. In this review, we summarize the results we obtained with different xCT-targeting vaccines. We show that, despite xCT being a self-antigen, vaccination was able to induce a humoral immune response that delayed primary tumor growth and strongly impaired pulmonary metastasis formation in mice challenged with tumorsphere-derived cells. Moreover, immunotargeting of xCT was able to increase CSC chemosensitivity to doxorubicin, suggesting that it may act as an adjuvant to chemotherapy. In conclusion, our approach based on the comparison of the transcriptome of tumorspheres and parental cells allowed us to identify a novel CSC-related target and to develop preclinical therapeutic approaches able to impact on CSC biology, and therefore, hampering tumor growth and dissemination.


Subject(s)
Amino Acid Transport System y+/immunology , Breast Neoplasms/immunology , Cancer Vaccines/immunology , Neoplastic Stem Cells/immunology , Amino Acid Transport System y+/metabolism , Animals , Cell Line, Tumor , Female , Humans , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/therapy , Mice, Inbred BALB C
4.
Oncoimmunology ; 7(3): e1408746, 2018.
Article in English | MEDLINE | ID: mdl-29399412

ABSTRACT

Aggressive forms of breast cancer, such as Her2+ and triple negative breast cancer (TNBC), are enriched in breast cancer stem cells (BCSC) and have limited therapeutic options. BCSC represent a key cellular reservoir for relapse, metastatic progression and therapeutic resistance. Their ability to resist common cytotoxic therapies relies on different mechanisms, including improved detoxification. The cystine-glutamate antiporter protein xCT (SLC7A11) regulates cystine intake, conversion to cysteine and subsequent glutathione synthesis, protecting cells against oxidative and chemical insults. Our previous work showed that xCT is highly expressed in tumorspheres derived from breast cancer cell lines and downregulation of xCT altered BCSC function in vitro and inhibited pulmonary metastases in vivo. We further strengthened these observations by developing a virus-like-particle (VLP; AX09-0M6) immunotherapy targeting the xCT protein. AX09-0M6 elicited a strong antibody response against xCT including high levels of IgG2a antibody. IgG isolated from AX09-0M6 treated mice bound to tumorspheres, inhibited xCT function as assessed by reactive oxygen species generation and decreased BCSC growth and self-renewal. To assess if AX09-0M6 impacts BCSC in vivo seeding, Her2+ TUBO-derived tumorspheres were injected into the tail vein of AX09-0M6 or control treated female BALB/c mice. AX09-0M6 significantly inhibited formation of pulmonary nodules. To evaluate its ability to impact metastases, AX09-0M6 was administered to mice with established subcutaneous 4T1 tumors. AX09-0M6 administration significantly hampered tumor growth and development of pulmonary metastases. These data show that a VLP-based immunization approach inhibits xCT activity, impacts BCSC biology and significantly reduces metastatic progression in preclinical models.

5.
FASEB J ; 28(1): 416-29, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24088817

ABSTRACT

Lactoferrin, a key component of innate immunity, is a cationic monomeric 80-kDa glycoprotein of the transferrin superfamily. Recombinant human lactoferrin, known as talactoferrin (TLF), induces a distinct functional maturation program in human dendritic cells (DCs) derived from peripheral blood monocytes. However, the receptors and molecular mechanisms involved in this induction have not been fully determined. By exploiting genome-wide transcription profiling of immature DCs, TNF-α- and IL-1ß-matured DCs (m-DCs), and TLF-matured DCs (TLF-DCs), we have detected a set of transcripts specific for m-DCs and one specific for TLF-DCs. Functional network reconstruction highlighted, as expected, the association of m-DC maturation with IL-1ß, TNF-α, and NF-κB, whereas TLF-DC maturation was associated with ERK and NF-κB. This involvement of ERK and NF-κB transduction factors suggests direct involvement of Toll-like receptors (TLRs) in TLF-induced maturation. We have used MyD88 inhibition and siRNA silencing TLRs on human DCs and mouse TLR-2-knockout cells, to show that TLF triggers the maturation of both human and mouse DCs through TLR-2 and TLR-4.


Subject(s)
Dendritic Cells/metabolism , Lactoferrin/pharmacology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Dendritic Cells/drug effects , Female , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 5/metabolism
6.
Br J Clin Pharmacol ; 75(5): 1231-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23116458

ABSTRACT

AIMS: To ascertain the tolerability profile of single and repeated oral doses of methanesulfonyl fluoride (MSF, SNX-001) in healthy aged subjects, and to determine the degree of erythrocyte acetylcholinesterase (AChE) inhibition induced by MSF after single and repeated oral doses. METHODS: To calculate properly the kinetics and the duration of AChE inhibition, the effects of MSF were also studied in rodents. These experiments suggested that MSF administered three times per week should provide safe and efficacious AChE inhibition. In a randomized placebo-controlled phase I study, 3.6 mg, 7.2 mg or 10.8 mg MSF were then orally administered to 27 consenting healthy volunteers (aged 50 to 72 years). After a single dose phase and a 1 week wash-out period, the subjects received the same doses three times per week for 2 weeks. RESULTS: Twenty-two out of the 27 subjects completed the study. Four patients withdrew due to adverse events (AEs) and one for non-compliance. Erythrocyte AChE was inhibited by a total of 33%, 46%, and 62% after 2 weeks of 3.6 mg, 7.2 mg and 10.8 mg MSF, respectively. No serious AEs occurred. The most frequent AEs were headache (27%), nausea (11%) and diarrhoea (8%). CONCLUSIONS: MSF proved to be well tolerated even with repeated oral dosing. It is estimated that MSF provided a degree of AChE inhibition that should effectively enhance memory. This molecule deserves to be tested for efficacy in a pilot randomized controlled study in patients with Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/pharmacokinetics , Sulfones/pharmacokinetics , Acetylcholinesterase/metabolism , Aged , Alzheimer Disease/metabolism , Animals , Cholinesterase Inhibitors/adverse effects , Cholinesterase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Double-Blind Method , Erythrocyte Membrane/enzymology , Female , Half-Life , Humans , Male , Middle Aged , Rats , Rats, Sprague-Dawley , Sulfones/adverse effects , Sulfones/pharmacology
7.
FASEB J ; 22(8): 2747-57, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18364398

ABSTRACT

Lactoferrin (LF) is an important protein component of the innate immune system that is broadly distributed within the body fluids. LF is endowed with multiple biological activities. Talactoferrin (TLF), a recombinant human LF, is in clinical development as an anticancer agent and is entering Phase III clinical trials. Here, we show that TLF induces the maturation of human dendritic cells (DCs) derived from monocytes. TLF, at physiologically relevant concentrations (100 microg/ml) up-regulates the expression of human leukocyte antigen (HLA) class II, CD83, CD80, and CD86 costimulatory molecule and CXCR4 and CCR7 chemokine receptors, acting primarily through the p38 MAPK signaling pathway. DCs matured by TLF displayed an enhanced release of IL-8 and CXCL10, as well as a significantly reduced production of IL-6, IL-10, and CCL20. They also display a reduced ability to take up antigen and increased capacity to trigger proliferation and release IFN-gamma in the presence of allogeneic human T cells. TLF-matured DCs are able to prime naive T cells to respond to KLH antigen and display a significantly increased capacity to present Flu-MA(58-66) peptide to HLA-A2-matched T cells. These data suggest that a key immunomodulatory function that may be mediated by TLF is to link the innate with adaptive immunity through DC maturation.


Subject(s)
Dendritic Cells/drug effects , Lactoferrin/pharmacology , Cell Differentiation/drug effects , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Immunity, Innate , In Vitro Techniques , Lactoferrin/immunology , Lymphocyte Activation/drug effects , Models, Biological , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
8.
Cancer Res ; 67(13): 6425-32, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17616703

ABSTRACT

We have previously shown that talactoferrin-alfa (TLF), a recombinant human lactoferrin, is an immunomodulatory protein that is active against implanted tumors, both as a single agent and in combination with chemotherapy. In this study, we show that talactoferrin is active against autochthonous tumors in a transgenic mouse line, which is more analogous to human cancers, and identify key mechanistic steps involved in the anticancer activity of oral TLF. BALB/c mice transgenic for the rat neu (ErbB2) oncogene (BALB-neuT) treated with oral TLF showed a significant delay in carcinogenesis, with 60% tumor protection relative to vehicle-treated mice at week 21. Oral TLF also showed tumor growth inhibition in wild-type BALB/c mice implanted with neu(+) mammary adenocarcinoma, with one third displaying a long-lasting or complete response. Oral TLF induces an increase in intestinal mucosal IFN-gamma production and an increase in Peyer's patch cellularity, including expansion of CD8(+) T lymphocytes and NKT cells, and the enhancement of CD8(+) T-cell cytotoxicity. In IFN-gamma knockout mice, there is an absence of the TLF-induced Peyer's patch cellularity, no expansion of CD8(+) T lymphocytes and NKT cells, and loss of TLF anticancer activity. TLF antitumor activity is also lost in mice depleted of CD8(+) T cells and in CD1 knockout mice, which lack NKT activity. Thus, the inhibition of distant tumors by oral TLF seems to be mediated by an IFN-gamma-dependent enhancement of CD8(+) T- and NKT cell activity initiated within the intestinal mucosa.


Subject(s)
Adenocarcinoma/metabolism , CD8-Positive T-Lymphocytes/metabolism , Interferon-gamma/physiology , Killer Cells, Natural/metabolism , Lactoferrin/pharmacology , Neoplasms/drug therapy , Receptor, ErbB-2/biosynthesis , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Female , Interferon-gamma/metabolism , Lactoferrin/administration & dosage , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Peyer's Patches/metabolism
9.
Hum Gene Ther ; 16(8): 937-46, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16076252

ABSTRACT

Systemic gene delivery using cationic liposome-DNA complexes (LDCs) has been shown to elicit potent antitumor activity in mice with tumor metastases to the lungs. However, intravenous gene delivery for treatment of established cancer has not been evaluated previously in a spontaneous, large animal model. We therefore evaluated the safety, toxicity, and efficacy of intravenous gene delivery, using LDCs in dogs with established tumor metastases. Twenty dogs with chemotherapy-resistant osteosarcoma metastases to the lungs received a series of intravenous infusions of cationic liposomes and plasmid DNA encoding the canine interleukin-2 (IL-2) cDNA. Effects of intravenous gene delivery on immune activation, clinical and hematologic parameters, tumor responses, and survival times were assessed. We found that slow intravenous administration of IL-2 LDCs resulted in detectable IL-2 transgene expression in lung tissues of dogs. Repeated intravenous infusions of LDCs were well tolerated by dogs with lung tumor metastases and elicited systemic immune activation, as reflected by fever, leukogram changes, monocyte activation, and increased natural killer cell activity. Three of 20 dogs experienced partial or complete regression of lung metastases after infusion of IL-2 LDCs. Overall survival times were significantly increased in treated dogs compared with historical control animals with the same stage of disease. We conclude that repeated intravenous infusion of LDCs in cancerbearing dogs is safe and well tolerated at low doses and may be capable of eliciting antitumor activity in some animals with advanced tumor metastases.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/pathology , DNA/administration & dosage , Gene Transfer Techniques , Interleukin-2/biosynthesis , Interleukin-2/genetics , Lung Neoplasms/secondary , Osteosarcoma/genetics , Osteosarcoma/secondary , Animals , Bone Neoplasms/therapy , Bone Neoplasms/veterinary , DNA/analysis , Dog Diseases/genetics , Dog Diseases/pathology , Dog Diseases/therapy , Dogs , Drug Resistance, Neoplasm , Gene Expression Profiling , Infusions, Intravenous , Killer Cells, Natural , Liposomes , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Lung Neoplasms/veterinary , Neoplasm Metastasis , Osteosarcoma/therapy , Osteosarcoma/veterinary , Plasmids , Survival Analysis , Transgenes
10.
Int J Cancer ; 111(3): 398-403, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15221967

ABSTRACT

In this work, we investigated the anticancer activity of orally administered recombinant human lactoferrin (rhLF) alone and in combination with chemotherapy in tumor-bearing mice. rhLF inhibited the growth of squamous cell carcinoma (O12) tumors in T cell-immunocompromised nu/nu mice by 80% when administered at 1,000 mg/kg (2.9 g/m2) by oral gavage twice daily for 8 days (p < 0.001). Similar activity was observed in syngeneic, immunocompetent BALB/c mice, where orally administered rhLF (1,000 mg/kg, 2.9 g/m2 once daily) halted the growth of mammary adenocarcinoma TUBO. Oral rhLF (200 mg/kg, 0.57 g/m2) was also used alone and in combination with cis-platinum (5 mg/kg) to treat head-and-neck squamous cell carcinoma in a syngeneic murine model. Monotherapy with oral rhLF or cis-platinum caused 61% or 66% tumor growth inhibition over placebo, respectively. Mice receiving both therapies showed 79% growth inhibition, a statistically significant improvement over each drug alone. We then demonstrated that administration of oral rhLF (300 mg/kg, 0.86 g/m2) to tumor-bearing or naive mice resulted in (i) significantly increased production of IL-18 in the intestinal tract, (ii) systemic NK cell activation and (iii) circulating CD8+ T-cell expansion. These data suggest that oral rhLF is an immunomodulatory agent active against cancer as a single agent and in combination chemotherapy, exerting its systemic effect through stimulation of IL-18 and other cytokines in the gut enterocytes. rhLF has been administered orally to 211 people without a single serious drug-related adverse event. Thus, rhLF shows promise as a safe and well-tolerated novel immunomodulatory anticancer agent.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/pathology , Cell Division/drug effects , Killer Cells, Natural/immunology , Lactoferrin/therapeutic use , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Cisplatin/therapeutic use , Drug Synergism , Female , Flow Cytometry , Humans , Killer Cells, Natural/drug effects , Lactoferrin/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Nude , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use
11.
Cancer Res ; 64(8): 2858-64, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15087404

ABSTRACT

The transforming rat Her-2/neu oncogene embedded into the genome of virgin transgenic BALB/c mice (BALB-neuT) provokes the development of an invasive carcinoma in each of their 10 mammary glands. i.m. vaccination with DNA plasmids coding for the extracellular and transmembrane domains of the protein product of the Her-2/neu oncogene started when mice already display multifocal in situ carcinomas temporarily halts neoplastic progression, but all mice develop a tumor by week 43. By contrast, progressive clearance of neoplastic lesions and complete protection of all 1-year-old mice are achieved when the same plasmids are electroporated at 10-week intervals. Pathological findings, in vitro tests, and the results from the immunization of both IFN-gamma and immunoglobulin gene knockout BALB-neuT mice, and of adoptive transfer experiments, all suggest that tumor clearance rests on the combination of antibodies and IFN-gamma-releasing T cells. These findings show that an appropriate vaccine effectively inhibits the progression of multifocal preneoplastic lesions.


Subject(s)
Genes, erbB-2/genetics , Immunotherapy, Active/methods , Immunotherapy, Adoptive/methods , Mammary Neoplasms, Experimental/therapy , Receptor, ErbB-2/immunology , Vaccines, DNA/administration & dosage , Animals , Antibody-Dependent Cell Cytotoxicity , Carcinoma in Situ/genetics , Carcinoma in Situ/immunology , Carcinoma in Situ/therapy , Electroporation/methods , Female , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Receptor, ErbB-2/genetics , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology
12.
Arch Otolaryngol Head Neck Surg ; 129(6): 618-22, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12810463

ABSTRACT

OBJECTIVES: To demonstrate that the combination of nonviral murine interleukin 2 (mIL-2) gene therapy and external-beam radiation therapy (XRT) have an enhanced therapeutic effect for the treatment of head and neck squamous cell carcinoma (HNSCC) in an orthotopic murine model and to elucidate the mechanism of action. METHODS: Randomized, controlled studies in the murine orthotopic model of HNSCC. Squamous cell carcinoma VII cells were injected into the floor of the mouth to establish tumors in immunocompetent mice. The intervention groups were treated with mIL-2, radiation therapy, empty plasmid, no treatment, combination mIL-2/XRT, and combination empty plasmid/XRT. Nonviral mIL-2 gene transfer was performed on days 5 and 9. The XRT was administered to the assigned groups 24 hours after first mIL-2 delivery. The mice were killed on day 13. Tumors and local lymph nodes were harvested and evaluated. Primary and secondary cytokine expression, cytotoxic T-lymphocyte activity, and apoptosis were assayed. RESULTS: The combination mIL-2/XRT demonstrated a significant increase in antitumor effects compared with single therapy or controls. Increased expression levels of primary and secondary cytokines were found in the group treated with mIL-2, and this effect was preserved when mIL-2 treatment was combined with XRT. Combination therapy significantly increased apoptosis compared with monotherapy. CONCLUSIONS: The present study demonstrates that combination mIL-2/XRT generates potent antitumor immune responses and significantly increases apoptosis in an orthotopic murine model of HNSCC. Further optimization of this strategy is warranted as well as consideration for human clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/therapy , Genetic Therapy/methods , Head and Neck Neoplasms/therapy , Interleukin-2/genetics , Interleukin-2/therapeutic use , Radiotherapy/methods , Animals , Apoptosis/physiology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/physiopathology , Combined Modality Therapy , Cytokines/biosynthesis , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/physiopathology , Interferon-gamma/biosynthesis , Mice , Models, Animal , T-Lymphocytes, Cytotoxic/physiology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...