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










Publication year range
1.
BMJ Open ; 13(8): e069858, 2023 08 07.
Article in English | MEDLINE | ID: mdl-37550029

ABSTRACT

INTRODUCTION: The purpose of TheShinISS-Vax|Flu study is to examine the association between influenza vaccines and adverse events requiring hospital admission or emergency care during the influenza vaccination campaigns 2021/2022 and 2022/2023 in Italy. METHODS AND ANALYSIS: This is a Self-Controlled Case Series multiregional study using linked routinely collected data from regional healthcare databases of the participating regions. Study participants will be persons aged ≥6 months, unvaccinated or who have received influenza vaccine during the influenza vaccination campaigns in the seasons 2021/2022 and 2022/2023 in Italy and who have experienced the outcome of interest for the first time during the study period (1 September 2021-30 June 2022 and 1 September 2022-30 June 2023 for the first and second vaccination campaigns, respectively). Risk periods will be specifically defined for each outcome and further subdivided into periods of 7 days. The exposures will be the first or second dose of the influenza vaccines administered during the two vaccination campaigns. Statistical analysis will be conducted separately for the data of the two campaigns. Exposure risk period will be compared with baseline risk period defined as any time of observation out of the risk periods. The modified SCCS method will be applied to handle event-dependent exposure and mortality and fitted using unbiased estimating equations to estimate relative incidences and excess of cases per 100 000 vaccinated by dose, age, sex and type of vaccine. Calendar period will be included as time-varying confounder in the model, where appropriate. ETHICS AND DISSEMINATION: The study received the approval from the National ethics committee for clinical trials of public research bodies and other national public institutions (PRE BIO CE n.0036723, 23/09/2022). Results will be published in peer-reviewed journals and reports in accordance with the publication policies of the Italian National Institute of Health and of the Italian Medicines Agency.


Subject(s)
Influenza Vaccines , Influenza, Human , Humans , Influenza Vaccines/therapeutic use , Influenza, Human/epidemiology , Vaccination , Immunization Programs , Italy , Observational Studies as Topic
2.
PLoS Med ; 19(7): e1004056, 2022 07.
Article in English | MEDLINE | ID: mdl-35900992

ABSTRACT

BACKGROUND: Myocarditis and pericarditis following the Coronavirus Disease 2019 (COVID-19) mRNA vaccines administration have been reported, but their frequency is still uncertain in the younger population. This study investigated the association between Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) mRNA vaccines, BNT162b2, and mRNA-1273 and myocarditis/pericarditis in the population of vaccinated persons aged 12 to 39 years in Italy. METHODS AND FINDINGS: We conducted a self-controlled case series study (SCCS) using national data on COVID-19 vaccination linked to emergency care/hospital discharge databases. The outcome was the first diagnosis of myocarditis/pericarditis between 27 December 2020 and 30 September 2021. Exposure risk period (0 to 21 days from the vaccination day, subdivided in 3 equal intervals) for first and second dose was compared with baseline period. The SCCS model, adapted to event-dependent exposures, was fitted using unbiased estimating equations to estimate relative incidences (RIs) and excess of cases (EC) per 100,000 vaccinated by dose, age, sex, and vaccine product. Calendar period was included as time-varying confounder in the model. During the study period 2,861,809 persons aged 12 to 39 years received mRNA vaccines (2,405,759 BNT162b2; 456,050 mRNA-1273); 441 participants developed myocarditis/pericarditis (346 BNT162b2; 95 mRNA-1273). Within the 21-day risk interval, 114 myocarditis/pericarditis events occurred, the RI was 1.99 (1.30 to 3.05) after second dose of BNT162b2 and 2.22 (1.00 to 4.91) and 2.63 (1.21 to 5.71) after first and second dose of mRNA-1273. During the [0 to 7) days risk period, an increased risk of myocarditis/pericarditis was observed after first dose of mRNA-1273, with RI of 6.55 (2.73 to 15.72), and after second dose of BNT162b2 and mRNA-1273, with RIs of 3.39 (2.02 to 5.68) and 7.59 (3.26 to 17.65). The number of EC for second dose of mRNA-1273 was 5.5 per 100,000 vaccinated (3.0 to 7.9). The highest risk was observed in males, at [0 to 7) days after first and second dose of mRNA-1273 with RI of 12.28 (4.09 to 36.83) and RI of 11.91 (3.88 to 36.53); the number of EC after the second dose of mRNA-1273 was 8.8 (4.9 to 12.9). Among those aged 12 to 17 years, the RI was of 5.74 (1.52 to 21.72) after second dose of BNT162b2; for this age group, the number of events was insufficient for estimating RIs after mRNA-1273. Among those aged 18 to 29 years, the RIs were 7.58 (2.62 to 21.94) after first dose of mRNA-1273 and 4.02 (1.81 to 8.91) and 9.58 (3.32 to 27.58) after second dose of BNT162b2 and mRNA-1273; the numbers of EC were 3.4 (1.1 to 6.0) and 8.6 (4.4 to 12.6) after first and second dose of mRNA-1273. The main study limitations were that the outcome was not validated through review of clinical records, and there was an absence of information on the length of hospitalization and, thus, the severity of the outcome. CONCLUSIONS: This population-based study of about 3 millions of residents in Italy suggested that mRNA vaccines were associated with myocarditis/pericarditis in the population younger than 40 years. According to our results, increased risk of myocarditis/pericarditis was associated with the second dose of BNT162b2 and both doses of mRNA-1273. The highest risks were observed in males of 12 to 39 years and in males and females 18 to 29 years vaccinated with mRNA-1273. The public health implication of these findings should be considered in the light of the proven mRNA vaccine effectiveness in preventing serious COVID-19 disease and death.


Subject(s)
COVID-19 Vaccines , COVID-19 , Myocarditis , Pericarditis , 2019-nCoV Vaccine mRNA-1273 , Adolescent , Adult , BNT162 Vaccine , COVID-19/diagnosis , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , Child , Female , Humans , Italy/epidemiology , Male , Myocarditis/chemically induced , Myocarditis/epidemiology , Pericarditis/chemically induced , Pericarditis/epidemiology , Product Surveillance, Postmarketing , SARS-CoV-2 , Vaccination/adverse effects , Young Adult
3.
Pharmaceuticals (Basel) ; 15(5)2022 Apr 25.
Article in English | MEDLINE | ID: mdl-35631352

ABSTRACT

Considering the clinical significance for myocarditis and pericarditis after immunization with mRNA COVID-19 vaccines, the present pharmacovigilance study aimed to describe these events reported with mRNA COVID-19 vaccines in the Vaccine Adverse Events Reporting System (VAERS). From 1990 to July 2021, the mRNA vaccines were the most common suspected vaccines related to suspected cases of myocarditis and/or pericarditis (myocarditis: N = 1,165; 64.0%; pericarditis: N = 743; 55.1%), followed by smallpox vaccines (myocarditis: N = 222; 12.2%; pericarditis: N = 200; 14.8%). We assessed all suspected cases through the case definition and classification of the Brighton Collaboration Group, and only definitive, probable, and possible cases were included in the analysis. Our findings suggested that myocarditis and pericarditis mostly involve young male, especially after the second dose with a brief time to onset. Nevertheless, this risk is lower (0.38/100,000 vaccinated people; 95% CI 0.36-0.40) than the risk of developing myocarditis after SARS-CoV-2 infection (1000-4000 per 100,000 people) and the risk of developing "common" viral myocarditis (1-10 per 100,000 people/year). Comparing with the smallpox vaccine, for which is already well known the association with myocarditis and pericarditis, our analysis showed a lower probability of reporting myocarditis (ROR 0.12, 95% CI 0.10-0.14) and pericarditis (ROR 0.06, 95% CI 0.05-0.08) following immunization with mRNA COVID-19 vaccines.

4.
Bioorg Med Chem ; 27(15): 3248-3253, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31208798

ABSTRACT

Tenatumomab is an anti-tenascin murine monoclonal antibody previously used in clinical trials for delivering radionuclides to tumors by both pre-targeting (biotinylated Tenatumomab within PAGRIT) and direct 131Iodine labeling approaches. Here we present the synthesis and in vitro characterization of three Tenatumomab conjugates to bifunctional chelating agents (NHS-DOTA, NCS-DOTA and NCS-DTPA). Results indicate ST8198AA1 (Tenatumomab-DOTAMA, derived by conjugation of NHS-DOTA), as the most promising candidate in terms of conjugation rate and yield, stability, antigen immunoreactivity and affinity. Labeling efficiency of the different chelators was investigated with a panel of cold metals indicating DOTAMA as the best chelator. Labeling of Tenatumomab-DOTAMA was then optimized with several metals and stability performed confirms suitability of this conjugate for further development. ST8198AA1 represents an improvement of the previous antibody forms because the labeling with radionuclides like 177Lu or 64Cu would allow theranostic applications in patients bearing tenascin expressing tumors.


Subject(s)
Heterocyclic Compounds, 1-Ring/pharmacology , Neoplasms/drug therapy , Tenascin/antagonists & inhibitors , Theranostic Nanomedicine , Dose-Response Relationship, Drug , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Molecular Structure , Neoplasms/genetics , Structure-Activity Relationship , Tenascin/genetics
5.
Oncotarget ; 9(11): 9766-9775, 2018 Feb 09.
Article in English | MEDLINE | ID: mdl-29515769

ABSTRACT

The clinical outcome of T-cell non-Hodgkin lymphoma (NHL) is poor and innovative treatments are needed. Tenascin-C is a large extracellular glycoprotein not expressed under physiological conditions, but overexpressed in cancer. Aim of the study was to evaluate tenascin-C expression within pathologic tissue of T-cell NHL and determine its clinical significance. We used an immunohistochemistry approach using the anti-tenascin-C monoclonal antibody Tenatumomab in 75 systemic T-cell NHL (including 72 mature and 3 precursor T-cell NHL), and 25 primary cutaneous T-cell NHL. Data were analyzed in terms of staining intensity, proportion of involved areas and histologic pattern, and results were correlated with clinical characteristics and outcome. Ninety-three percent of the cases were tenascin-C positive and 59% of systemic diseases were characterized by a predominant involvement (>50%). Stromal expression was detected in all the cases while vascular and vascular plus cytoplasmic expression was present in 49% and 23%. The constant overexpression of the tenascin-C gene was observed in two independent publicly available T-cell NHL gene expression datasets. In conclusions, tenascin-C represents an attractive target that sets the rationale to investigate the therapeutic activity of radiolabeled Tenatumomab in T-cell NHL.

6.
Oncotarget ; 8(14): 22590-22605, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28186982

ABSTRACT

The oxidized version of Avidin, known as AvidinOX, was previously shown to link to tissue proteins upon injection or nebulization, thus becoming a stable receptor for biotinylated therapeutics. AvidinOX is currently under clinical investigation to target radioactive biotin to inoperable tumor lesions (ClinicalTrials.gov NCT02053324). Presently, we show that the anti-ErbB2 monoclonal antibodies Trastuzumab and Pertuzumab can be chemically biotinylated while maintaining their biochemical and biological properties. By using several and diverse experimental conditions, we show that when AvidinOX is conjugated to tumor cells, low antibody concentrations of biotinylated Trastuzumab (bTrast) or Pertuzumab (bPert) prevent internalization of ErbB2, induce endoplasmic reticulum stress, cell cycle arrest and apoptosis leading to inhibition of proliferation and ErbB2 signaling. Moreover, we found that the treatment is able to induce down-modulation of ErbB2 thus bypassing the known resistance of this receptor to degradation. Interestingly, we show that AvidinOX anchorage is a way to counteract agonistic activities of Trastuzumab and Pertuzumab. Present data are in agreement with previous observations from our group indicating that the engagement of the Epidermal Growth Factor Receptor (EGFR) by AvidinOX-bound biotinylated Cetuximab or Panitumumab, leads to potent tumor inhibition both in vitro and in animal models. All results taken together encourage further investigation of AvidinOX-based treatments with biotinylated antibodies directed to the members of the EGFR family.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Avidin/metabolism , Breast Neoplasms/drug therapy , Receptor, ErbB-2/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/administration & dosage , Avidin/chemistry , Biomarkers, Tumor/metabolism , Biotinylation , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Female , Humans , Receptor, ErbB-2/immunology , Signal Transduction , Trastuzumab/administration & dosage , Tumor Cells, Cultured
7.
Oncotarget ; 7(1): 914-28, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26575422

ABSTRACT

For locally advanced and metastatic head and neck squamous cell carcinoma (HNSCC), the current clinical use of Cetuximab in chemo/radiotherapy protocols is often associated to severe systemic toxicity. Here we report in vitro data in human FaDu pharynx SCC cells, showing that inactive concentrations of biotinylated Cetuximab (bCet) become active upon anchorage to AvidinOX on the surface of tumor cells. AvidinOX-anchored bCet induces apoptosis and DNA damage as well as specific inhibition of signaling, degradation and abrogation of nuclear translocation of EGFR. In the mouse model of FaDu cancer, we show that intra-tumor injection of AvidinOX allows anti-tumor activity of an otherwise inactive, intraperitoneally delivered, low dose bCet. Consistently with in vitro data, in vivo tumor inhibition is associated to induction of apoptosis, DNA damage and reduced angiogenesis. AvidinOX is under clinical investigation for delivering radioactive biotin to inoperable tumors (ClinicalTrials.gov NCT02053324) and present data support its use for the local treatment of HNSCC in combination with systemic administration of low dose bCet.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Avidin/administration & dosage , Biotinylation , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cetuximab/administration & dosage , Dose-Response Relationship, Drug , ErbB Receptors/metabolism , Female , Head and Neck Neoplasms/blood supply , Head and Neck Neoplasms/pathology , Humans , Immunoblotting , Immunohistochemistry , Mice, Nude , Neovascularization, Pathologic/prevention & control , Treatment Outcome , Tumor Burden/drug effects
8.
Mol Oncol ; 9(9): 1760-72, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26119717

ABSTRACT

An awesome number of experimental and clinical evidences indicate that constitutive activation of the Met oncogenic receptor plays a critical role in the progression of cancer toward metastasis and/or resistance to targeted therapies. While mutations are rare, the common mechanism of Met activation is overexpression, either by gene amplification ('addiction') or transcriptional activation ('expedience'). In the first instance ligand-independent kinase activation plays a major role in sustaining the transformed phenotype. Anti-Met antibodies directed against the receptor binding site behave essentially as ligand (Hepatocyte Growth Factor, HGF) antagonists and are ineffective to counteract ligand-independent activation. The monovalent chimeric MvDN30 antibody fragment, PEGylated to extend its half-life, binds the fourth IPT domain and induces 'shedding' of the Met extracellular domain, dramatically reducing both the number of receptors on the surface and their phosphorylation. Downstream signaling is thus inhibited, both in the absence or in the presence of the ligand. In vitro, MvDN30 is a strong inhibitor not only of ligand-dependent invasive growth, sustained by both paracrine and autocrine HGF, but notably, also of ligand-independent growth of 'Met-addicted' cells. In immunocompromised mice, lacking expression of Hepatocyte Growth Factor cross-reacting with the human receptor - thus providing, by definition, a model of 'ligand-independent' Met activation - PEGylated MvDN30 impairs growth of Met 'addicted' human gastric carcinoma cells. In a Met-amplified patient-derived colo-rectal tumor (xenopatient) MvDN30-PEG overcomes the resistance to EGFR targeted therapy (Cetuximab). The PEGylated MvDN30 is thus a strong candidate for targeting tumors sustained by ligand-independent Met oncogenic activation.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Female , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/pharmacology , Immunoglobulin Fragments/therapeutic use , Mice , Neoplasms/metabolism , Neoplasms/pathology , Polyethylene Glycols/chemistry , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/metabolism
9.
Oncotarget ; 5(19): 9239-55, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25238453

ABSTRACT

Lung cancer, as well as lung metastases from distal primary tumors, could benefit from aerosol treatment. Unfortunately, because of lung physiology, clearance of nebulized drugs is fast, paralleled by unwanted systemic exposure. Here we report that nebulized AvidinOX can act as an artificial receptor for biotinylated drugs. In nude and SCID mice with advanced human KRAS-mutated A549 metastatic lung cancer, pre-nebulization with AvidinOX enables biotinylated Cetuximab to control tumor growth at a dose lower than 1/25,000 the intravenous effective dose. This result correlates with a striking, specific and unpredictable effect of AvidinOX-anchored biotinylated Cetuximab, as well as Panitumumab, observed on a panel of tumor cell lines, leading to inhibition of dimerization and signalling, blockade of endocytosis, induction of massive lysosomal degradation and abrogation of nuclear translocation of EGFR. Excellent tolerability, together with availability of pharmaceutical-grade AvidinOX and antibodies, will allow rapid clinical translation of the proposed therapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Avidin/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Active Transport, Cell Nucleus/drug effects , Administration, Inhalation , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab , Endocytosis/drug effects , ErbB Receptors/metabolism , Humans , Lysosomes/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Panitumumab , Protein Multimerization/drug effects , Signal Transduction/drug effects
10.
PLoS One ; 6(6): e21075, 2011.
Article in English | MEDLINE | ID: mdl-21701578

ABSTRACT

We recently reported that the oxidized avidin, named AvidinOX®, resides for weeks within injected tissues as a consequence of the formation of Schiff's bases between its aldehyde groups and tissue protein amino groups. We also showed, in a mouse pre-clinical model, the usefulness of AvidinOX for the delivery of radiolabeled biotin to inoperable tumors. Taking into account that AvidinOX is the first oxidized glycoprotein known to chemically link to injected tissues, we tested in the mouse a panel of additional oxidized glycoproteins, with the aim of investigating the phenomenon. We produced oxidized ovalbumin and mannosylated streptavidin which share with avidin glycosylation pattern and tetrameric structure, respectively and found that neither of them linked significantly to cells in vitro nor to injected tissues in vivo, despite the presence of functional aldehyde groups. The study, extended to additional oxidized glycoproteins, showed that the in vivo chemical conjugation is a distinctive property of the oxidized avidin. Relevance of the high cationic charge of avidin into the stable linkage of AvidinOX to tissues is demonstrated as the oxidized acetylated avidin lost the property. Plasmon resonance on matrix proteins and cellular impedance analyses showed in vitro that avidin exhibits a peculiar interaction with proteins and cells that allows the formation of highly stable Schiff's bases, after oxidation.


Subject(s)
Avidin/metabolism , 3T3 Cells , Animals , Cell Line, Tumor , Chickens , Glycoproteins/metabolism , Glycosylation , Humans , Male , Mice , Microscopy, Confocal , Ovalbumin/metabolism , Protein Binding , Streptavidin/metabolism , Surface Plasmon Resonance
11.
Basic Clin Pharmacol Toxicol ; 109(3): 145-55, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21426491

ABSTRACT

We recently described an oxidized avidin variant, named AvidinOX(®) , which is a product that chemically links to tissue proteins while maintaining the capacity to uptake intravenously administered biotin. Such product proved to be successful in targeting radionuclide therapy in a mouse model of inoperable breast cancer. Here, we show that the uptake of a single or multiple doses of biotin (up to five times), by the tissue-bound AvidinOX(®) , is stable for 2 weeks. Taking into account that oxidized avidin is the first chemically reactive protein to be proposed for clinical use, we evaluated its tolerability, immunogenicity and mutagenicity. Present in vitro data indicate that AvidinOX(®) (up to 10 µg/5 × 10(5) cells) does not affect cell viability or proliferation of PC3 human prostate cancer or 3T3 mouse fibroblast cell lines as well as primary mouse spleen cells. Safety pharmacology and toxicology studies were conducted using AvidinOX(®) up to the highest concentration compatible with its solubility (about 12 mg/mL), representing four times the product concentration intended for human use, and in the maximum administrable volume compatible with each study system. The intramuscular administration in rat and monkey induced a moderate to strong inflammatory response particularly after a second administration and consistently with the induction of an immune response. Interestingly, the intramuscular administration of AvidinOX(®) to rodents and monkeys exhibiting very high anti-avidin antibody titres was well tolerated with no systemic symptoms of any kind. Intravenous administration of AvidinOX(®) , performed to mimic an accidental injection of the dose intended for a local administration (15 µL of 3.3 mg/mL solution), showed significant localization of the product into the spleen not associated with uptake of the radiolabelled biotin intravenously injected after 24 hr, thus suggesting rapid inactivation. No mutagenic activity was induced by oxidized avidin in prokaryotic and eukaryotic cells. Overall, the present data indicate that AvidinOX(®) is well tolerated in rodents and non-human primates, thus supporting its clinical use within protocols of radionuclide therapy of inoperable tumour lesions.


Subject(s)
Avidin/pharmacology , Avidin/toxicity , Biotin/administration & dosage , Indium Radioisotopes , Radiopharmaceuticals/pharmacology , Radiopharmaceuticals/toxicity , 3T3 Cells , Animals , Apoptosis/drug effects , Avidin/immunology , Avidin/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Kidney/drug effects , Kidney/pathology , Liver/drug effects , Liver/pathology , Macaca fascicularis , Male , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Mutagenicity Tests/methods , Radiopharmaceuticals/immunology , Radiopharmaceuticals/pharmacokinetics , Rats , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Toxicity Tests/methods
12.
J Biol Chem ; 285(46): 36149-57, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20833723

ABSTRACT

Met, the high affinity receptor for hepatocyte growth factor, is one of the most frequently activated tyrosine kinases in human cancer and a validated target for cancer therapy. We previously developed a mouse monoclonal antibody directed against the extracellular portion of Met (DN-30) that induces Met proteolytic cleavage (receptor "shedding") followed by proteasome-mediated receptor degradation. This translates into inhibition of hepatocyte growth factor/Met-mediated biological activities. However, DN-30 binding to Met also results in partial activation of the Met kinase due to antibody-mediated receptor homodimerization. To safely harness the therapeutic potential of DN-30, its shedding activity must be disassociated from its agonistic activity. Here we show that the DN-30 Fab fragment maintains high affinity Met binding, elicits efficient receptor shedding and down-regulation, and does not promote kinase activation. In Met-addicted tumor cell lines, DN-30 Fab displays potent cytostatic and cytotoxic activity in a dose-dependent fashion. DN-30 Fab also inhibits anchorage-independent growth of several tumor cell lines. In mouse tumorigenesis assays using Met-addicted carcinoma cells, intratumor administration of DN-30 Fab or systemic delivery of a chemically stabilized form of the same molecule results in reduction of Met phosphorylation and inhibition of tumor growth. These data provide proof of concept that monovalency unleashes the full therapeutic potential of the DN-30 antibody and point at DN-30 Fab as a promising tool for Met-targeted therapy.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Antibodies, Monoclonal/pharmacology , Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/immunology , Animals , Antibodies, Anti-Idiotypic/immunology , Antibodies, Anti-Idiotypic/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibody Affinity/immunology , Apoptosis/drug effects , Binding, Competitive , Blotting, Western , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , HCT116 Cells , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fab Fragments/pharmacology , Immunoprecipitation , Mice , Mice, Nude , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Cancer Biother Radiopharm ; 25(5): 563-70, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20863248

ABSTRACT

Hen egg white avidin is increasingly used in the clinic as part of multifactor treatments such as pretargeted radionuclide therapy of cancer or as an antidote of biotinylated drugs. Taking into account that naturally occurring human antiavidin antibodies (HAVA) are common in humans, the present work investigates avidin immunogenicity as part of risk/benefit evaluations. Sera from 139 oncology patients naive to avidin were confirmed to exhibit HAVA with lognormally distributed titers. HAVA were boosted after avidin treatment, with no correlation with the avidin dose or with the basal titer. No antibody-related clinical symptoms were observed in 21 HAVA-positive patients treated with avidin. In mouse models, high mouse antiavidin antibody titers, induced to simulate the worst human condition, neither reduced the biotin uptake of intratissue-injected avidin nor affected the capacity of intravenously injected avidin to clear a biotinylated drug from circulation. In both models the avidin treatment was well tolerated. Results indicate that avidin immunogenicity does not affect its safety and efficacy, thus encouraging its further use in clinical applications.


Subject(s)
Antibodies/adverse effects , Avidin/administration & dosage , Avidin/immunology , Biotin/pharmacokinetics , Neoplasms/therapy , Radioimmunotherapy/adverse effects , Animals , Antibodies/blood , Avidin/therapeutic use , Humans , Mice , Mice, Inbred BALB C , Models, Animal , Radioimmunotherapy/methods , Tissue Distribution
14.
J Biomed Biotechnol ; 2009: 921434, 2009.
Article in English | MEDLINE | ID: mdl-20130784

ABSTRACT

Avidin is a glycoprotein from hen egg white that binds biotin with very high affinity. Here we describe OXavidin, a product containing aldehyde groups, obtained by ligand-assisted sugar oxidation of avidin by sodium periodate. OXavidin chemically reacts with cellular and tissue proteins through Schiff's base formation thus residing in tissues for weeks while preserving the biotin binding capacity. The long tissue residence of OXavidin as well as that of OXavidin/biotinylated agent complex occurs in normal and neoplastic tissues and immunohistochemistry shows a strong and homogenous stromal localization. Once localized in tissue/tumor, OXavidin becomes an "artificial receptor" for intravenous injected biotin allowing tumor targeting with biotinylated therapeutics like radioisotopes or toxins. Moreover, present data also suggest that OXavidin might be useful for the homing of biotinylated cells. Overall, OXavidin exhibits a remarkable potential for many different therapeutic applications.


Subject(s)
Avidin/pharmacokinetics , Breast Neoplasms/drug therapy , Drug Delivery Systems/methods , Immunohistochemistry/methods , Animals , Avidin/chemistry , Avidin/metabolism , Avidin/therapeutic use , Biotin/metabolism , Cell Line, Tumor , Female , Half-Life , Humans , Mice , Neoplasm Transplantation , Periodic Acid , Streptavidin/pharmacokinetics
15.
BMC Biotechnol ; 7: 70, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17945015

ABSTRACT

BACKGROUND: There is much evidence that tumor cells elicit a humoral immune response in patients. In most cases, the presence of antibodies in peripheral blood is detected only in small proportion of patients with tumors overexpressing the corresponding antigen. In the present study, we analyzed the significance of local humoral response provided by tumor-infiltrating lymphocytes in breast cancer patients. METHODS: The ability of a patient's immune system to produce specific antibodies inside tumor tissue, capable of recognizing tumor cells, was explored through analysis of the oligoclonality of antibodies derived from tumor-infiltrating lymphocytes and construction of a series of recombinant antibody libraries in scFv format, derived from breast tumor-infiltrating B lymphocytes. These libraries and one from peripheral blood lymphocytes of a single breast cancer patient were panned against three purified surface tumor antigens, such as CEA, MUC1 and ED-B domain, and against intact MCF7 breast carcinoma cells. RESULTS: Application of novel display vector, pKM19, allowed isolation of a large panel of breast cancer-specific antibodies against known tumor antigens, as well as against breast carcinoma cells. Reactivity of novel scFvs was confirmed by ELISA, immunohistochemistry, fluorescence staining and flow cytometry. We demonstrated that seven of ten primary breast tumor specimens, obtained using discarded surgical material, could be exploited as an appropriate source for generation of phage display libraries, giving highly specific antitumor antibodies which recognize heterologous tumor cells. CONCLUSION: Local humoral immune response within tumor tissue in breast cancer patients frequently has an oligoclonal character. Efficient selection of specific antitumor antibodies from recombinant antibody libraries, derived from such oligoclonal tumor-infiltrated B lymphocytes, indicates the presence of natural immune response against tumor antigens in these patients. The described method is very promising for development of antitumor antibodies, potentially useful for diagnostic and therapeutic approaches.


Subject(s)
B-Lymphocytes/immunology , Breast Neoplasms/immunology , Epitope Mapping/methods , Immunoglobulins/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Cell Line, Tumor , Humans
16.
Clin Cancer Res ; 12(7 Pt 1): 2191-6, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16609034

ABSTRACT

ST2146biot is a biotinylated anti-tenascin monoclonal antibody (mAb) to be used for Pretargeted Antibody Guided Radioimmunotherapy (PAGRIT) of solid tumors. In vivo biodistribution studies of (125)I-labeled ST2146biot were done in nude mice transplanted with human HT-29 colon carcinoma and/or human U-118MG glioblastoma cells characterized for low and high tenascin expression, respectively. In vitro results show that ST2146 retains immunoreactivity upon biotinylation, in contrast to other anti-tenascin mAbs. In vivo biodistribution of ST2146 shows specific tumor accumulation up to 10 days after the i.v. injection, with no relevant differences between biotinylated and nonbiotinylated ST2146. A dose of 4 microg/mouse saturates the low tenascin-expressing human colon carcinoma HT-29, whereas the high tenascin-expressing human glioblastoma U-118MG seems to be saturated at a ST2146biot dose between 320 and 640 microg/mouse. The percentage of injected dose per gram of tumor ranges from 10% to 30%, corresponding to an amount of ST2146biot/g of tumor of approximately 400 ng/g and >200 microg/g for HT-29 and U-118MG, respectively. Tumor to normal organs uptake ratios are between 15 and 60, confirming high tumor selectivity of ST2146biot despite its cross-reactivity with the tenascin expressed at low level in the normal mouse organs. The ST2146biot localization data are substantially confirmed even when both low and high tenascin-expressing tumors are implanted in the same animal. To our knowledge, the absolute amount of ST2146biot, specifically localized in xenotransplanted human tumors, is the highest thus far described and supports the clinical use of this mAb in PAGRIT(R).


Subject(s)
Adenocarcinoma/metabolism , Antibodies, Monoclonal/pharmacokinetics , Antibody Specificity , Breast Neoplasms/metabolism , Colonic Neoplasms/metabolism , Glioblastoma/metabolism , Tenascin/immunology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Antibodies, Monoclonal/chemistry , Biotinylation , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Disease Models, Animal , Drug Screening Assays, Antitumor , Glioblastoma/immunology , Glioblastoma/pathology , Humans , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Radioimmunotherapy/methods , Structure-Activity Relationship , Tenascin/biosynthesis , Transplantation, Heterologous , Xenograft Model Antitumor Assays
17.
BMC Cancer ; 6: 41, 2006 Feb 24.
Article in English | MEDLINE | ID: mdl-16504122

ABSTRACT

BACKGROUND: CEA is a tumor-associated antigen abundantly expressed on several cancer types, including those naturally refractory to chemotherapy. The selection and characterization of human anti-CEA single-chain antibody fragments (scFv) is a first step toward the construction of new anticancer monoclonal antibodies designed for optimal blood clearance and tumor penetration. METHODS: The human MA39 scFv, selected for its ability to recognize a CEA epitope expressed on human colon carcinomas, was first isolated from a large semi-synthetic ETH-2 antibody phage library, panned on human purified CEA protein. Subsequently, by in vitro mutagenesis of a gene encoding for the scFv MA39, a new library was established, and new scFv antibodies with improved affinity towards the CEA cognate epitope were selected and characterized. RESULTS: The scFv MA39 antibody was affinity-maturated by in vitro mutagenesis and the new scFv clone, E8, was isolated, typed for CEA family member recognition and its CEACAM1, 3 and 5 shared epitope characterized for expression in a large panel of human normal and tumor tissues and cells. CONCLUSION: The binding affinity of the scFv E8 is in a range for efficient, in vivo, antigen capture in tumor cells expressing a shared epitope of the CEACAM1, 3 and 5 proteins. This new immunoreagent meets all criteria for a potential anticancer compound: it is human, hence poorly or not at all immunogenic, and it binds selectively and with good affinity to the CEA epitope expressed by metastatic melanoma and colon and lung carcinomas. Furthermore, its small molecular size should provide for efficient tissue penetration, yet give rapid plasma clearance.


Subject(s)
Carcinoembryonic Antigen/chemistry , Immunoglobulin Variable Region/chemistry , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Antibody Specificity , Antineoplastic Agents/pharmacology , Bacteria/metabolism , Biotinylation , Blotting, Western , Cell Line, Tumor , Colonic Neoplasms/metabolism , DNA/chemistry , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Flow Cytometry , Gene Library , Humans , Immunoglobulin Fragments/chemistry , Immunohistochemistry , Kinetics , Lung Neoplasms/metabolism , Melanoma/metabolism , Microscopy, Confocal , Molecular Sequence Data , Mutagenesis , Neoplasm Metastasis , Peptide Library , Protein Structure, Tertiary , Sensitivity and Specificity , Surface Plasmon Resonance , Transfection
18.
J Chromatogr A ; 1107(1-2): 182-91, 2006 Feb 24.
Article in English | MEDLINE | ID: mdl-16414054

ABSTRACT

Using phage display technology, a 22-mer peptide was selected as a ligand with unique specificity for the murine monoclonal ST2146 antibody that recognizes the EGF repeats region of the human tumor-associated antigen tenascin-C. This peptide, synthesized in an 8-branched form to enhance its binding properties, is useful in replacing the native antigen in the affinity and immunoreactivity characterization of the ST2146 antibody and its biotinylated derivatives. Affinity resins, prepared by immobilizing the mimotope or its shorter 10-mer binding unit on a chromatographic support, were able to capture ST2146 directly from the hybridoma supernatant, with antibody recovery and host cell protein (HCP) reduction similar to or better than protein A sorbent, a purity degree exceeding 95%, and full recovery of antibody activity. The affinity constants of both peptides, as determined by frontal analysis of broad-zone elution affinity chromatography and BiaCore measurements, were very similar and included in a range suitable for affinity ligands. Column capacity, determined by applying a large excess of purified ST2146 to 1 mL of column bed volume, was close to 50 mg/mL for both resins. These matrices retain their ST2146 binding properties after various treatments, including sanitization, thus indicating very high stability in terms of ligand leakage and degradation. Moreover, the short form shows higher enzymatic stability, thus proving more suitable as ligand for ST2146 affinity purification.


Subject(s)
Affinity Labels/chemistry , Antibodies, Monoclonal/immunology , Bacteriophages/chemistry , Peptides/chemistry , Tenascin/immunology , Affinity Labels/isolation & purification , Amino Acid Sequence , Antibodies, Monoclonal/isolation & purification , Blotting, Western , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Molecular Sequence Data , Peptides/immunology , Peptides/isolation & purification , Surface Plasmon Resonance
19.
Clin Cancer Res ; 11(19 Pt 2): 7137s-7145s, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16203813

ABSTRACT

PURPOSE: In the pretargeted antibody-guided radioimmunotherapy (PAGRIT) system, the combined use of two different antibodies directed against the same tumor antigen could represent a valid approach for improving tumor targeting and therapeutic efficacy. We developed a novel monoclonal antitenascin antibody, ST2485, and studied its biochemical and functional properties by in vitro and in vivo assays. We then investigated the first of the three-step therapy combining ST2485 with another antitenascin antibody, ST2146, previously described, to increase accumulation of biotinylated antibodies at the tumor site. EXPERIMENTAL DESIGN: Studies of immunoreactivity, affinity, immunohistochemistry, and biodistribution in xenograft model were carried out on ST2485. Analysis of the ST2485 and ST2146 combination was preliminary carried out by ELISA and BiaCore tests and then by in vivo distribution studies after administration of the radiolabeled biotinylated antibodies, followed by a chase with avidin as clearing agent. RESULTS: ST2485 was found to be a suitable antibody for therapeutic applications. Indeed, for its behavior in all tests, it was comparable with ST2146 and better than BC2, an antibody already used for clinical trials. The additivity of ST2146 and ST2485 in tenascin C binding, shown by in vitro tests, was confirmed by biodistribution studies in a xenograft model where tumor localization of the antibodies was near the sum of each antibody alone, with a tumor-to-blood ratio higher than 24. CONCLUSION: The results reported in this study suggest that a monoclonal antitenascin antibody mixture can improve tumor targeting. This strategy could represent progress for therapeutic approaches such as PAGRIT.


Subject(s)
Antibodies/chemistry , Neoplasms, Experimental/therapy , Radioimmunotherapy/methods , Tenascin/chemistry , Tenascin/immunology , Amino Acid Sequence , Animals , Antibodies/pharmacology , Antibodies, Monoclonal/chemistry , Antibody Specificity , Antigens, Neoplasm/chemistry , Biotinylation , Cell Line, Tumor , Clinical Trials as Topic , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Humans , Immunohistochemistry , Kinetics , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Neoplasm Transplantation , Peptides/chemistry , Protein Structure, Tertiary , Radioimmunodetection/methods , Recombinant Proteins/chemistry , Streptavidin/chemistry , Time Factors , Tissue Distribution
20.
Hum Mutat ; 20(5): 368-74, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12402334

ABSTRACT

In this study we examined a series of 52 patients belonging to hereditary nonpolyposis colorectal cancer (HNPCC) or HNPCC-related families, all who had previously tested negative for mismatch repair (MMR) gene point mutations. Southern blot mutational screening of MLH1 and MSH2 genes was carried out with the aim of detecting large genomic rearrangements and of identifying the molecular mechanisms underlying the inactivation of the MMR genes. Three patients had abnormal restriction patterns and were found to carry distinct MLH1 internal deletions. Long-range PCRs identified the loss of DNA tracts spanning exon 6 (about 2.4 kb in proband A-AV20 and 0.8 kb in proband A-PD5) and exon 3 (about 2.5 kb in proband R-RM2). In A-AV20 the breakpoints occurred into identical 33-bp regions in introns 5 and 6 and a mechanism of classical Alu-mediated homologous recombination was evident. Also, in patient A-PD5 the breakpoints were located in these introns, but without direct involvement of repetitive sequences. In patient R-RM2 the breakpoints were located within repetitive L1 elements with poor homology in intron 2 and 3 and the rearranged allele was characterized by a complex insertion deletion (delCCinsACATAGTA), giving rise to a palindromic CTTAACATAGTATGTTAAG sequence in proximity of the fusion site. This study confirms that genomic rearrangements are an important component of the spectrum of MMR mutations. Although Alu repeats are likely to be implicated in the majority of cases, different molecular mechanisms may also be responsible for the observed MLH1 intragenic deletions. In particular, HNPCC resulting from L1-mediated recombination has been identified as a novel mechanism for MMR inactivating mutation.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Neoplasm Proteins/genetics , Sequence Deletion , Adaptor Proteins, Signal Transducing , Adult , Alleles , Base Sequence , Carrier Proteins , Family Health , Genome, Human , Humans , Molecular Sequence Data , MutL Protein Homolog 1 , Nuclear Proteins , Sequence Alignment , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...