Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Oral Oncol ; 148: 106635, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37988837

RESUMO

OBJECTIVES: Adenoid cystic carcinoma (ACC) is a rare type of cancer that typically arises from glandular tissues, most commonly in the salivary glands. Although relatively rare, it represents a serious clinical issue as the management of the disease is highly complex being the only therapeutic options represented by invasive surgery and/or radiotherapy. In the present study, we have explored the potential of galectin-3 binding protein (LGALS3BP) as a novel target for antibody-drug conjugate (ADC) therapy in ACC. MATERIALS AND METHODS: RNAseq was conducted on a panel of 10 ACC patient-derived xenografts (PDX)s tissues and 6 normal salivary glands to analyze LGALS3BP gene expression. Protein expression was assessed in ACC PDX and primary tumor tissues using immunohistochemistry. Anti-LGALS3BP ADC named 1959-sss/DM4, was tested in high LGALS3BP expressing ACC PDX model ST1502B. RESULTS: RNAseq analysis revealed that LGALS3BP expression was highly expressed in ACC PDX tissues compared to normal salivary gland tissues. As evaluated by immunohistochemical analysis, LGALS3BP protein was found to be heterogeneously expressed in 10 ACC PDX and in tumor tissues derived from a cohort of 37 ACC patients. Further, treatment with 1959-sss/DM4 ADC led to durable tumor growth inhibition (TGI) in 100% of animals without observed toxicity. CONCLUSIONS: Our study provides strong evidence that LGALS3BP is a promising therapeutic target for ACC, warranting further expedited preclinical and clinical investigation.


Assuntos
Antígenos de Neoplasias , Biomarcadores Tumorais , Carcinoma Adenoide Cístico , Neoplasias das Glândulas Salivares , Animais , Humanos , Biomarcadores Tumorais/antagonistas & inibidores , Carcinoma Adenoide Cístico/tratamento farmacológico , Modelos Animais de Doenças , Neoplasias das Glândulas Salivares/tratamento farmacológico , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Idoso , Idoso de 80 Anos ou mais , Ensaios Antitumorais Modelo de Xenoenxerto , Camundongos
2.
Nat Med ; 29(12): 3149-3161, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38066208

RESUMO

The glial environment influences neurological disease progression, yet much of our knowledge still relies on preclinical animal studies, especially regarding astrocyte heterogeneity. In murine models of traumatic brain injury, beneficial functions of proliferating reactive astrocytes on disease outcome have been unraveled, but little is known regarding if and when they are present in human brain pathology. Here we examined a broad spectrum of pathologies with and without intracerebral hemorrhage and found a striking correlation between lesions involving blood-brain barrier rupture and astrocyte proliferation that was further corroborated in an assay probing for neural stem cell potential. Most importantly, proteomic analysis unraveled a crucial signaling pathway regulating this astrocyte plasticity with GALECTIN3 as a novel marker for proliferating astrocytes and the GALECTIN3-binding protein LGALS3BP as a functional hub mediating astrocyte proliferation and neurosphere formation. Taken together, this work identifies a therapeutically relevant astrocyte response and their molecular regulators in different pathologies affecting the human cerebral cortex.


Assuntos
Astrócitos , Células-Tronco Neurais , Humanos , Camundongos , Animais , Astrócitos/patologia , Proteômica , Encéfalo , Sistema Nervoso Central
3.
Cell Death Discov ; 9(1): 400, 2023 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-37898642

RESUMO

HER-3 (also known as ErbB-3) is a human epidermal growth factor receptor tyrosine kinases family member, and its expression in CRC (colorectal cancer) tissues was previously associated with poor prognosis. In this study, HER-3 expression was analyzed by immunohistochemistry in two cohorts of early and advanced metastatic CRC patients. The first cohort included 180 patients diagnosed with CRC in absence of lymph nodes or distant metastases (Stage I and Stage II), while the second was obtained from 53 advanced metastatic CRC patients who developed synchronous (SM) and metachronous (MM) liver metastases. In the first early-stage CRC cohort, 86 out of 180 (47.8%) tumors showed membranous expression of HER-3, with a mean percentage of positive tumor cells of 25.7%; conversely, in advanced metastatic CRC primary tumors, HER-3 was detected in all specimens, with a mean percentage of positive tumor cells of 76.1%. Kaplan-Meier curves showed that in the advanced metastatic CRC group, patients with HER-3high tumors had a significantly lower Cancer-Specific Survival (CSS) rate compared to patients with HER-3low tumors (p = 0.021). Importantly, this worse CSS rate was observed only in the MM subgroup of patients with HER-3high tumors (p = 0.002). Multivariate analysis confirmed that high HER-3 expression represents a significant and strong risk factor for death in patients developing MM liver metastases (Hazard Ratio = 64.9; 95% Confidence Interval, 4.7-886.6; p = 0.002). In addition, using a specific anti-HER-3 antibody-drug conjugate, named EV20/MMAF, we showed that HER-3 + CRC cells can be efficiently targeted in vitro and in vivo. Overall, this study confirms that surface HER-3 is highly expressed in CRC and reveals that HER-3 expression increases in metastatic CRC patients compared to early stage. Importantly, the results suggest that HER-3 has a prognostic and therapeutic value in patients developing MM liver metastases.

4.
J Pharm Biomed Anal ; 235: 115642, 2023 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-37586307

RESUMO

The present study focuses on the development and validation of an HPLC-DAD methodology for the detection of a potent chemotherapeutic agent, Maytansinoid Ravtansine (DM4), and its metabolite, S-methyl-DM4 (S-Me-DM4), in plasma samples. Methodologically, after a simple protein precipitation with acetonitrile and after drying 1 mL of supernatant, the sample (suspended with N,N-Dimethylacetamide, DMA) was directly analyzed by HPLC under isocratic elution using a mobile phase comprising milliQ water and methanol (25:75, v:v), both acidified with 0.1 % v:v formic acid. Employing a flow rate of 1.0 mL/min and a reversed-phase GraceSmart RP18 column thermostated at 40 °C, we achieved complete resolution and separation of DM4 and S-Me-DM4 within 13 min. The optimized injection volume of 20 µL and the wavelength set at 254 nm were utilized for quantitative analyses. Rigorous validation has not only ensured its reliability and reproducibility but has also addressed potential limitations associated with methodological inconsistency. The limit of detection and quantification of the method were 0.025 and 0.06 µg/mL for both the analytes, respectively. The calibration curve showed a good linearity in the range 0.06-20 µg/mL. For both analytes, the intraday precision and trueness were 2.3-8.2 % and -1.1 to 3.1 %, respectively, while the interday values were 0.7-10.1 % and -10.4 to 7.5 %, respectively. The developed methodology enables the concurrent determination and quantification of free DM4 and its metabolite, free S-Me-DM4, making it a valuable tool for assessing the pharmacokinetics and pharmacodynamics of DM4-based therapies. In addition, the procedure was successfully applied to analyse the presence of free DM4 or its metabolite, free S-Me-DM4, in human plasma samples spiked with the 1959-sss/DM4 antibody-drug conjugate (ADC). The utilization of the herein validated methodology allowed to confirm the presence of these analytes, thereby providing insights into their potential release from the ADC structure.


Assuntos
Maitansina , Humanos , Reprodutibilidade dos Testes , Cromatografia Líquida de Alta Pressão/métodos , Preparações Farmacêuticas
5.
Mol Pharm ; 20(6): 3241-3248, 2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-37191353

RESUMO

Galectin-3 binding protein (Gal-3BP) is a glycoprotein that is overexpressed and secreted by several cancers and has been implicated as a marker of both tumor progression and poor prognosis in melanoma, non-small cell lung cancer, head and neck squamous cell carcinoma, and breast cancer. The expression of Gal-3BP by a variety of neoplasms makes it an enticing target for both diagnostics and therapeutics, including immuno-positron emission tomography (immunoPET) probes and antibody-drug conjugates (ADCs). Herein, we report the development, in vitro characterization, and in vivo evaluation of a pair of Gal-3BP-targeting radioimmunoconjugates for 89Zr-immunoPET. A humanized anti-Gal-3BP antibody, 1959, and its corresponding ADC, 1959-sss/DM4 (DM4 = ravtansine), were modified with desferrioxamine (DFO) to yield DFO-1959 and DFO-1959-sss/DM4 immunoconjugates bearing 1-2 DFO/monoclonal antibody. Both DFO-modified immunoconjugates retained their affinity for Gal-3BP in enzyme-linked immunosorbent assay experiments. The chelator-bearing antibodies were radiolabeled with zirconium-89 (t1/2 ≈ 3.3 d) to produce radioimmunoconjugates ─ [89Zr]Zr-DFO-1959 and [89Zr]Zr-DFO-1959-sss/DM4 ─ with high specific activity (>444 MBq/mg, >12 mCi/mg) and stability (>80% intact after 168 h in human serum at 37 °C). In mice bearing subcutaneous Gal-3BP-secreting A375-MA1 xenografts, [89Zr]Zr-DFO-1959 clearly delineated tumor tissue, reaching a maximum tumoral activity concentration (54.8 ± 15.8%ID/g) and tumor-to-background contrast (tumor-to-blood = 8.0 ± 4.6) at 120 h post-injection. The administration of [89Zr]Zr-DFO-1959 to mice bearing subcutaneous Gal-3BP-expressing melanoma patient-derived xenografts produced similarly promising results. [89Zr]Zr-DFO-1959 and [89Zr]Zr-DFO-1959-sss/DM4 exhibited nearly identical pharmacokinetic profiles in the mice bearing A375-MA1 tumors, though the latter produced higher uptake in the spleen and kidneys. Both [89Zr]Zr-DFO-1959 and [89Zr]Zr-DFO-1959-sss/DM4 effectively visualized Gal-3BP-secreting tumors in murine models of melanoma. These results suggest that both probes could play a role in the clinical imaging of Gal-3BP-expressing malignancies, particularly as companion theranostics for the identification of patients likely to respond to Gal-3BP-targeted therapeutics such as 1959-sss/DM4.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Imunoconjugados , Neoplasias Pulmonares , Melanoma , Animais , Humanos , Camundongos , Linhagem Celular Tumoral , Desferroxamina/química , Galectina 3 , Imunoconjugados/química , Tomografia por Emissão de Pósitrons/métodos , Zircônio/química
6.
Clin Exp Med ; 23(7): 3689-3700, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37162650

RESUMO

Glycoprotein 90K, encoded by the interferon-stimulated gene LGALS3BP, displays broad antiviral activity. It reduces HIV-1 infectivity by interfering with Env maturation and virion incorporation, and increases survival of Influenza A virus-infected mice via antiviral innate immune signaling. Its antiviral potential in SARS-CoV-2 infection remains largely unknown. Here, we analyzed the expression of 90K/LGALS3BP in 44 hospitalized COVID-19 patients at multiple levels. We quantified 90K protein concentrations in serum and PBMCs as well as LGALS3BP mRNA levels. Complementary, we analyzed two single cell RNA-sequencing datasets for expression of LGALS3BP in respiratory specimens and PBMCs from COVID-19 patients. Finally, we analyzed the potential of 90K to interfere with SARS-CoV-2 infection of HEK293T/ACE2, Calu-3 and Caco-2 cells using authentic virus. 90K protein serum concentrations were significantly elevated in COVID-19 patients compared to uninfected sex- and age-matched controls. Furthermore, PBMC-associated concentrations of 90K protein were overall reduced by SARS-CoV-2 infection in vivo, suggesting enhanced secretion into the extracellular space. Mining of published PBMC scRNA-seq datasets uncovered monocyte-specific induction of LGALS3BP mRNA expression in COVID-19 patients. In functional assays, neither 90K overexpression in susceptible cell lines nor exogenous addition of purified 90K consistently inhibited SARS-CoV-2 infection. Our data suggests that 90K/LGALS3BP contributes to the global type I IFN response during SARS-CoV-2 infection in vivo without displaying detectable antiviral properties in vitro.


Assuntos
COVID-19 , Humanos , Animais , Camundongos , Células CACO-2 , Células HEK293 , Leucócitos Mononucleares , SARS-CoV-2 , Antivirais , RNA Mensageiro , Antígenos de Neoplasias , Biomarcadores Tumorais
7.
Mol Oncol ; 17(8): 1460-1473, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37195369

RESUMO

Glioblastoma multiforme (GBM) is a lethal disease characterized by an overall survival of about 1 year, making it one of the most aggressive tumours, with very limited therapeutic possibilities. Specific biomarkers for early diagnosis as well as innovative therapeutic strategies are urgently needed to improve the management of this deadly disease. In this work, we demonstrated that vesicular galectin-3-binding protein (LGALS3BP), a glycosylated protein overexpressed in a variety of human malignancies, is a potential GBM disease marker and can be efficiently targeted by a specific antibody-drug conjugate (ADC). Immunohistochemical analysis on patient tissues showed that LGALS3BP is highly expressed in GBM and, compared with healthy donors, the amount of vesicular but not total circulating protein is increased. Moreover, analysis of plasma-derived extracellular vesicles from mice harbouring human GBM revealed that LGALS3BP can be used for liquid biopsy as a marker of disease. Finally, an ADC targeting LGALS3BP, named 1959-sss/DM4, specifically accumulates in tumour tissue, producing a potent and dose-dependent antitumor activity. In conclusion, our work provides evidence that vesicular LGALS3BP is a potential novel GBM diagnostic biomarker and therapeutic target deserving further preclinical and clinical validation.


Assuntos
Neoplasias Encefálicas , Vesículas Extracelulares , Glioblastoma , Imunoconjugados , Humanos , Animais , Camundongos , Glioblastoma/diagnóstico , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Biomarcadores Tumorais/metabolismo , Imunoconjugados/farmacologia , Imunoconjugados/uso terapêutico , Vesículas Extracelulares/metabolismo , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Antígenos de Neoplasias/metabolismo
8.
Clin Exp Med ; 23(1): 151-155, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35076790

RESUMO

Coronavirus disease 2019 (COVID-19) has quickly turned into a health, financial and societal problem globally. The complex pathogenesis of severe acute respiratory syndrome coronavirus centers on the unpredictable clinical progression of the disease, which may evolve abruptly and results in critical and life-threatening clinical complications. Effective laboratory biomarkers that can classify patients according to risk of progression to severe disease are essential for ensuring timely treatment. Gal-3BP is a human secreted protein with innate immune functions, which is upregulated in viral infections, promotes inflammation and has been shown to induce IL-6 expression. In this study, Gal-3BP plasma levels were measured retrospectively in a cohort of 84 hospitalized COVID-19 patients. These were classified as having either "non-severe" or "severe" disease. Compared to healthy controls, Gal-3BP plasma levels were markedly increased in COVID-19 patients (P < 0.0001). Moreover, the levels were higher in severe than in non-severe patients (P < 0.05). As expected, patients with severe disease had plasma levels of IL-6 higher than patients with non-severe disease (P < 0.01). In non-severe disease patients, Gal-3BP levels collected at a late stage (13.3 + 5.7 days after the first positive PCR result) were significantly lower than those collected at an early stage (4.2 + 2.9 days form the first positive PCR result). Larger prospective analyses are needed to strength our understanding of the prognostic utility of Gal-3BP in COVID-19 patients.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Interleucina-6 , Estudos Prospectivos , Estudos Retrospectivos
9.
Sci Rep ; 12(1): 17047, 2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-36220879

RESUMO

COVID-19 is the global pandemic that affected our population in the past 2 years. Considerable research has been done to better understand the pathophysiology of this disease and to identify new therapeutic targets, especially for severe cases. Galectin-3 (Gal-3) is a receptor present at the surface of different cell types, namely epithelial and inflammatory cells, which has been described as a severity marker in COVID-19. The activation of Gal-3 through its binding protein (Gal-3BP) is directly linked to the production of pro-inflammatory cytokines that contribute for the cytokine storm (CS) observed in severe COVID-19 patients. Here, we show that D2, a recombinant fragment of the lectin-binding region of Gal-3BP was able to stimulate the expression of IL-6 in colon and lung epithelial cell lines in ß-galactoside dependent manner. We further show that D2-induced IL-6 augmentation was reduced by the anti-Gal-3BP monoclonal antibody 1959. Our data confirm and extend prior findings of Gal-3BP mediated IL-6 induction, enlightening the potential of its antibody-mediated s blockage for the prevention and treatment of CS and severe disease in COVID-19 patients.


Assuntos
Tratamento Farmacológico da COVID-19 , SARS-CoV-2 , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Proteínas de Transporte , Linhagem Celular , Síndrome da Liberação de Citocina , Citocinas/metabolismo , Galectina 3/metabolismo , Humanos , Interleucina-6/metabolismo
10.
Int J Mol Sci ; 23(13)2022 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-35806317

RESUMO

Galectin-3 binding protein (Gal-3BP) is a multifunctional glycoprotein involved in cell-cell and cell-matrix interactions known to be upregulated in cancer and various viral infections, including HIV-1, HCV, and SARS-CoV-2, with a key role in regulating the antiviral immune response. Studies have identified a direct correlation between circulating levels of Gal-3BP and the severity of disease and/or disease progression for some viral infections, including SARS-CoV-2, suggesting a role of Gal-3BP in these processes. Due to Gal-3BP's complex biology, the molecular mechanisms underlying its role in viral diseases have been only partially clarified. Gal-3BP induces the expression of interferons (IFNs) and proinflammatory cytokines, including interleukin-6 (IL-6), mainly interacting with galectin-3, targeting the TNF receptor-associated factors (TRAF-6 and TRAF-3) complex, thus having a putative role in the modulation of TGF-ß signaling. In addition, an antiviral activity of Gal-3BP has been ascribed to a direct interaction of the protein with virus components. In this review, we explored the role of Gal-3BP in viral infections and the relationship between Gal-3BP upregulation and disease severity and progression, mainly focusing on SARS-CoV-2. Augmented knowledge of Gal-3BP's role in virus infections can be useful to evaluate its possible use as a prognostic biomarker and as a putative target to block or attenuate severe disease.


Assuntos
COVID-19 , Viroses , Antivirais , Galectina 3/metabolismo , Humanos , SARS-CoV-2
11.
J Transl Med ; 19(1): 405, 2021 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-34565385

RESUMO

The lectin galactoside-binding soluble 3 binding protein (LGALS3BP) is a secreted, hyperglycosylated protein expressed by the majority of human cells. It was first identified as cancer and metastasis associated protein, while its role in innate immune response upon viral infection remains still to be clarified. Since its discovery dated in early 90 s, a large body of literature has been accumulating highlighting both a prognostic and functional role for LGALS3BP in cancer. Moreover, data from our group and other have strongly suggested that this protein is enriched in cancer-associated extracellular vesicles and may be considered a promising candidate for a targeted therapy in LGALS3BP positive cancers. Here, we extensively reviewed the literature relative to LGALS3BP role in cancer and its potential value as a therapeutic target.


Assuntos
Galectina 3 , Neoplasias , Antígenos de Neoplasias , Biomarcadores Tumorais , Proteínas de Transporte , Linhagem Celular Tumoral , Galectina 3/metabolismo , Glicoproteínas/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Prognóstico
12.
Pharmaceutics ; 13(4)2021 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-33918158

RESUMO

HER-3 is becoming an attractive target for antibody-drug conjugate (ADC)-based therapy. Indeed, this receptor and its ligands are found to be overexpressed in several malignancies, and re-activation of its downstream signaling axis is known to play a critical role in modulating the sensitivity of targeted therapeutics in different tumors. In this study, we generated a novel ADC named EV20/NMS-P945 by coupling the anti-HER-3 antibody EV20 with a duocarmycin-like derivative, the thienoindole (TEI) NMS-P528, a DNA minor groove alkylating agent through a peptidic cleavable linker. This ADC showed target-dependent cytotoxic activity in vitro on several tumor cell lines and therapeutic activity in mouse xenograft tumor models, including those originating from pancreatic, prostatic, head and neck, gastric and ovarian cancer cells and melanoma. Pharmacokinetics and toxicological studies in monkeys demonstrated that this ADC possesses a favorable terminal half-life and stability and it is well tolerated. These data support further EV20/NMS-P945 clinical development as a therapeutic agent against HER-3-expressing malignancies.

13.
Oncol Rep ; 45(2): 776-785, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33416143

RESUMO

Liver cancer (LC) is an aggressive disease with a markedly poor prognosis. Therapeutic options are limited, and, until recently the only FDA­approved agent for first­line treatment of patients with LC was the multi­kinase inhibitor sorafenib, which exhibits limited activity and an increased overall survival (OS) of only 3 months over placebo. Therefore, the development of alternative therapeutic molecules for the treatment of LC is an urgent medical need. Antibody­drug conjugates (ADCs) are an emerging class of novel anticancer agents, which have been developed recently for the treatment of malignant conditions, including LC, and are being studied in preclinical and clinical settings. Our group has recently generated an ADC [EV20/monomethyl auristatin F (MMAF)] by coupling the HER3 targeting antibody (EV20) to MMAF via a non­cleavable maleimidocaproyl linker. This ADC was revealed to possess potent therapeutic activity in melanoma and breast carcinoma. In the present study, using western blot and flow cytometric analysis, it was reported that HER­3 receptor was highly expressed in LC and activated by its ligand NRG­1ß in a panel of LC cell lines, thus indicating that this receptor may serve as a suitable target for ADC therapy. A novel ADC [EV20­sss­valine­citrulline (vc)/MMAF] was generated, in which the cytotoxic payload MMAF was site­specifically coupled to an engineered variant of EV20 via a vc cleavable linker. Cytotoxicity assays were performed to investigate in vitro antitumor activity of EV20­sss­vc/MMAF and it was compared to EV20/MMAF, which revealed only modest activity in LC.EV20­sss­vc/MMAF exhibited a significant cell killing activity in several LC cell lines. Additionally, in vivo xenograft experiments revealed that EV20­sss­vc/MMAF inhibited growth of LC tumors. The present data indicated that EV20­sss­vc/MMAF is a worthy candidate for the treatment of HER­3 positive LC.


Assuntos
Imunoconjugados/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Oligopeptídeos/farmacologia , Receptor ErbB-3/antagonistas & inibidores , Idoso , Idoso de 80 Anos ou mais , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Imunoconjugados/uso terapêutico , Fígado/patologia , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Oligopeptídeos/uso terapêutico , Receptor ErbB-3/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Transl Med ; 19(1): 24, 2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-33407599

RESUMO

BACKGROUND: Benign prostatic hyperplasia (BPH) is the most common urologic disease among elderly men. The diagnosis of BPH is usually driven by lower urinary tract symptoms (LUTS) that can significantly affect patients' quality of life. This phase II prospective, randomized double-blinded, placebo-controlled study aimed to determine the efficacy and safety of a novel whole tomato-based food supplement on LUTS of patients diagnosed with BPH. METHODS: Forty consecutive patients with histologically proved BPH were randomized 1:1 to receive daily for 2 months a sachet (5 g) of a newly developed whole tomato food supplement (WTFS) (treatment = Group A) or placebo (Group B). Patients were asked to fill the International Prostatic Symptom Score (IPSS) questionnaire before and after treatment. RESULTS: All but 1 patient in Group B successfully completed the scheduled regimen. No side effects were recorded. Unlike placebo, treatment significantly reduced (P < 0.0002) LUTS since mean IPSS decreased from 9.05 ± 1.15 to 7.15 ± 1.04 (paired t-test, two-tailed P-value < 0.001), and improved life quality (P < 0.0001). A trend toward a reduction of total PSA levels was observed in WTFS treated patients (8.98 ng/mL ± 1.52 vs 6.95 ± 0.76, P = 0.065), with changes being statistically significant only in the subgroup of patients with baseline levels above 10 ng/mL (18.5 ng/mL ± 2.7 vs 10.3 ± 2.1, P = 0.009). CONCLUSIONS: The new WTFS may represent a valid option for the treatment of symptomatic BPH patients. Unlike pharmacological treatments, the supplement is side effects free and highly accepted among patients.


Assuntos
Hiperplasia Prostática , Solanum lycopersicum , Sistema Urinário , Idoso , Suplementos Nutricionais , Humanos , Hiperplasia , Masculino , Estudos Prospectivos , Hiperplasia Prostática/complicações , Hiperplasia Prostática/tratamento farmacológico , Qualidade de Vida , Resultado do Tratamento
15.
Cancers (Basel) ; 12(10)2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-33076448

RESUMO

Neuroblastoma is the most common extra-cranial solid tumor in infants and children, which accounts for approximately 15% of all cancer-related deaths in the pediatric population. New therapeutic modalities are urgently needed. Antibody-Drug Conjugates (ADC)s-based therapy has been proposed as potential strategy to treat this pediatric malignancy. LGALS3BP is a highly glycosylated protein involved in tumor growth and progression. Studies have shown that LGALS3BP is enriched in extracellular vesicles (EV)s derived by most neuroblastoma cells, where it plays a critical role in preparing a favorable tumor microenvironment (TME) through direct cross talk between cancer and stroma cells. Here, we describe the development of a non-internalizing LGALS3BP ADC, named 1959-sss/DM3, which selectively targets LGALS3BP expressing neuroblastoma. 1959-sss/DM3 mediated potent therapeutic activity in different types of neuroblastoma models. Notably, we found that treatments were well tolerated at efficacious doses that were fully curative. These results offer preclinical proof-of-concept for an ADC targeting exosomal LGALS3BP approach for neuroblastomas.

16.
Anal Bioanal Chem ; 412(27): 7659-7667, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32875368

RESUMO

The research and the individuation of tumour markers in biological fluids are currently one of the main tools to support diagnosis, prognosis, and monitoring of the therapeutic response in oncology. Although the identification of tumour markers in asymptomatic patients is crucial for early diagnosis, its application is still limited by the relatively low sensitivity and the complexity of existing methods (i.e. ELISA, mass spectrometry). We developed an easy, fast, and ultrasensitive surface-enhanced Raman scattering (SERS)-based system, for the detection and quantitation of the LGALS3BP (90K) biomarker that was used as a model, based on the development of antibody-functionalized nanostructured gold surfaces. The detection system was effective for the ultrasensitive detection and characterization of samples of different biochemical compositions. In conclusion, this work could provide the foundation for the development of a medical diagnostic device with the highest predictive power when compared with the methods currently used in cancer diagnostics.


Assuntos
Anticorpos Imobilizados/química , Antígenos de Neoplasias/sangue , Biomarcadores Tumorais/sangue , Nanoestruturas/química , Análise Espectral Raman/instrumentação , Antígenos de Neoplasias/análise , Biomarcadores Tumorais/análise , Desenho de Equipamento , Ouro/química , Humanos , Limite de Detecção , Neoplasias/sangue , Análise Espectral Raman/métodos
17.
Int J Mol Sci ; 21(15)2020 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-32752132

RESUMO

In recent years, antibody-drug conjugates (ADCs) have become promising antitumor agents to be used as one of the tools in personalized cancer medicine. ADCs are comprised of a drug with cytotoxic activity cross-linked to a monoclonal antibody, targeting antigens expressed at higher levels on tumor cells than on normal cells. By providing a selective targeting mechanism for cytotoxic drugs, ADCs improve the therapeutic index in clinical practice. In this review, the chemistry of ADC linker conjugation together with strategies adopted to improve antibody tolerability (by reducing antigenicity) are examined, with particular attention to ADCs approved by the regulatory agencies (the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA)) for treating cancer patients. Recent developments in engineering Immunoglobulin (Ig) genes and antibody humanization have greatly reduced some of the problems of the first generation of ADCs, beset by problems, such as random coupling of the payload and immunogenicity of the antibody. ADC development and clinical use is a fast, evolving area, and will likely prove an important modality for the treatment of cancer in the near future.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Imunoconjugados/uso terapêutico , Neoplasias/tratamento farmacológico , Anticorpos Monoclonais/imunologia , Antineoplásicos/imunologia , Humanos , Imunoconjugados/imunologia , Neoplasias/imunologia , Neoplasias/patologia
18.
EMBO Mol Med ; 12(5): e11498, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32329582

RESUMO

Despite impressive clinical benefit obtained with anti-HER2-targeted therapies, in advances stages, especially in the metastatic setting, HER2-positive tumors remain incurable. Therefore, it is important to develop novel strategies to fight these tumors, especially when they become resistant to available therapies. We show here that the anti-HER3 antibody-drug conjugate EV20/MMAF exerted potent anti-tumoral properties against several models of primary resistance and secondary resistance to common anti-HER2 available therapies, including trastuzumab, lapatinib, neratinib, and trastuzumab-emtansine. HER3 was expressed in these HER2+ breast cancer cells and knockdown experiments demonstrated that HER3 expression was required for the action of EV20/MMAF. In mice injected with trastuzumab-resistant HER2+ cells, a single dose of EV20/MMAF caused complete and long-lasting tumor regression. Mechanistically, EV20/MMAF bound to cell surface HER3 and became internalized to the lysosomes. Treatment with EV20/MMAF caused cell cycle arrest in mitosis and promoted cell death through mitotic catastrophe. These findings encourage the clinical testing of EV20/MMAF for several indications in the HER2+ cancer clinic, including situations in which HER2+ tumors become refractory to approved anti-HER2 therapies.


Assuntos
Antineoplásicos , Neoplasias da Mama , Imunoconjugados , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Imunoconjugados/farmacologia , Camundongos , Receptor ErbB-2 , Trastuzumab/farmacologia , Trastuzumab/uso terapêutico
19.
J Exp Clin Cancer Res ; 38(1): 191, 2019 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-31088502

RESUMO

BACKGROUND: The very aggressive nature and low survival rate of pancreatic ductal adenocarcinoma (PDAC) dictates the necessity to find novel efficacious therapies. Recent evidence suggests that phosphoinositide 3-kinase (PI3K) and 3-phosphoinositide-dependent protein kinase 1 (PDK1) are key effectors of oncogenic KRAS in PDAC. Herein, we report the role and mechanism of action of PDK1, a protein kinase of the AGC family, in PDAC. METHODS: PDAC cell lines were treated with selective PDK1 inhibitors or transfected with specific PDK1-targeting siRNAs. In vitro and in vivo assays were performed to investigate the functional role of PDK1 in PDAC. Specifically, anchorage-dependent and anchorage-independent growth was assessed in PDAC cells upon inhibition or downregulation of PDK1. Detailed investigation of the effect of PDK1 inhibition/downregulation on specific signalling pathways was also performed by Western blotting analysis. A xenograft tumour mouse model was used to determine the effect of pharmacological inhibition of PDK1 on PDAC cells growth in vivo. RESULTS: Treatment with specific inhibitors of PDK1 impaired anchorage-dependent and anchorage-independent growth of pancreatic cancer cell lines, as well as pancreatic tumour growth in a xenograft model. Mechanistically, inhibition or downregulation of PDK1 resulted in reduced activation of the serum/glucocorticoid regulated kinase family member 3 and subsequent reduced phosphorylation of its target N-Myc downstream regulated 1. Additionally, we found that combination of sub-optimal concentrations of inhibitors selective for PDK1 and the class IB PI3K isoform p110γ inhibits pancreatic cancer cell growth and colonies formation more potently than each single treatment. CONCLUSIONS: Our data indicate that PDK1 is a suitable target for therapeutic intervention in PDAC and support the clinical development of PDK1 inhibitors for PDAC.


Assuntos
Proteínas Quinases Dependentes de 3-Fosfoinositídeo/antagonistas & inibidores , Antineoplásicos/farmacologia , Neoplasias Pancreáticas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/genética , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/metabolismo , Animais , Biomarcadores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Fosfatidilinositol 3-Quinases/metabolismo , RNA Interferente Pequeno/genética , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Control Release ; 294: 176-184, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30553852

RESUMO

Galectin-3-binding protein (Gal-3BP) has been identified as a cancer and metastasis-associated, secreted protein that is expressed by the large majority of cancers. The present study describes a special type of non-internalizing antibody-drug-conjugates that specifically target Gal-3BP. Here, we show that the humanized 1959 antibody, which specifically recognizes secreted Gal-3BP, selectively localized around tumor but not normal cells. A site specific disulfide linkage with thiol-maytansinoids to unpaired cysteine residues of 1959, resulting in a drug-antibody ratio of 2, yielded an ADC product, which cured A375m melanoma bearing mice. ADC products based on the non-internalizing 1959 antibody may be useful for the treatment of several human malignancies, as the cognate antigen is abundantly expressed and secreted by several cancers, while being present at low levels in most normal adult tissues.


Assuntos
Antígenos de Neoplasias/imunologia , Biomarcadores Tumorais/imunologia , Imunoconjugados/uso terapêutico , Neoplasias/terapia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Imunoconjugados/farmacocinética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...