Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 54(8): 1772-1787.e9, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34289378

RESUMO

As substantial constituents of the multiple myeloma (MM) microenvironment, pro-inflammatory macrophages have emerged as key promoters of disease progression, bone destruction, and immune impairment. We identify beta-2-microglobulin (ß2m) as a driver in initiating inflammation in myeloma-associated macrophages (MAMs). Lysosomal accumulation of phagocytosed ß2m promotes ß2m amyloid aggregation in MAMs, resulting in lysosomal rupture and ultimately production of active interleukin-1ß (IL-1ß) and IL-18. This process depends on activation of the NLRP3 inflammasome after ß2m accumulation, as macrophages from NLRP3-deficient mice lack efficient ß2m-induced IL-1ß production. Moreover, depletion or silencing of ß2m in MM cells abrogates inflammasome activation in a murine MM model. Finally, we demonstrate that disruption of NLRP3 or IL-18 diminishes tumor growth and osteolytic bone destruction normally promoted by ß2m-induced inflammasome signaling. Our results provide mechanistic evidence for ß2m's role as an NLRP3 inflammasome activator during MM pathogenesis. Moreover, inhibition of NLRP3 represents a potential therapeutic approach in MM.


Assuntos
Amiloide/metabolismo , Mieloma Múltiplo/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Macrófagos Associados a Tumor/metabolismo , Microglobulina beta-2/metabolismo , Animais , Células Cultivadas , Humanos , Inflamação/imunologia , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Lisossomos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Fagocitose/imunologia , Transdução de Sinais/imunologia , Microambiente Tumoral/imunologia , Macrófagos Associados a Tumor/imunologia , Microglobulina beta-2/genética
2.
Int J Mol Sci ; 25(11)2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38892436

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest of human malignancies and carries an exceptionally poor prognosis. It is mostly driven by multiple oncogenic alterations, with the highest mutation frequency being observed in the KRAS gene, which is a key oncogenic driver of tumorogenesis and malignant progression in PDAC. However, KRAS remained undruggable for decades until the emergence of G12C mutation specific KRAS inhibitors. Despite this development, this therapeutic approach to target KRAS directly is not routinely used for PDAC patients, with the reasons being the rare presence of G12C mutation in PDAC with only 1-2% of occurring cases, modest therapeutic efficacy, activation of compensatory pathways leading to cell resistance, and absence of effective KRASG12D or pan-KRAS inhibitors. Additionally, indirect approaches to targeting KRAS through upstream and downstream regulators or effectors were also found to be either ineffective or known to cause major toxicities. For this reason, new and more effective treatment strategies that combine different therapeutic modalities aiming at achieving synergism and minimizing intrinsic or adaptive resistance mechanisms are required. In the current work presented here, pancreatic cancer cell lines with oncogenic KRAS G12C, G12D, or wild-type KRAS were treated with specific KRAS or SOS1/2 inhibitors, and therapeutic synergisms with concomitant MEK inhibition and irradiation were systematically evaluated by means of cell viability, 2D-clonogenic, 3D-anchorage independent soft agar, and bioluminescent ATP assays. Underlying pathophysiological mechanisms were examined by using Western blot analyses, apoptosis assay, and RAS activation assay.


Assuntos
Neoplasias Pancreáticas , Inibidores de Proteínas Quinases , Proteínas Proto-Oncogênicas p21(ras) , Humanos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/radioterapia , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Linhagem Celular Tumoral , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/radioterapia , Carcinoma Ductal Pancreático/terapia , Transdução de Sinais/efeitos dos fármacos , Apoptose , Mutação , Proliferação de Células/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo
3.
Cancer Immunol Immunother ; 70(1): 107-121, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32651619

RESUMO

Bromo- and extra-terminal domain (BET) inhibitors represent potential therapeutic approaches in solid and hematological malignancies that are currently analyzed in several clinical trials. Additionally, BET are involved in the epigenetic regulation of immune responses by macrophages and dendritic cells (DCs), that play a central role in the regulation of immune responses, indicating that cancer treatment with BET inhibitors can promote immunosuppressive effects. The aim of this study was to further characterize the effects of selective BET inhibition by JQ1 on DC maturation and DC-mediated antigen-specific T-cell responses. Selective BET inhibition by JQ1 impairs LPS-induced DC maturation and inhibits the migrational activity of DCs, while antigen uptake is not affected. JQ1-treated DCs show reduced ability to induce antigen-specific T-cell proliferation. Moreover, antigen-specific T cells co-cultured with JQ1-treated DCs exhibit an inactive phenotype and reduced cytokine production. JQ1-treated mice show reduced immune responses in vivo to sublethal doses of LPS, characterized by a reduced white blood cell count, an immature phenotype of splenic DCs and T cells and lower blood levels of IL-6. In our study, we demonstrate that selective BET inhibition by JQ1, a drug currently tested in clinical trials for malignant diseases, has profound effects on DC maturation and DC-mediated antigen-specific T-cell responses. These immunosuppressive effects can result in the induction of possible infectious side effects in cancer treatments. In addition, based on our results, these compounds should not be used in combinatorial regimes using immunotherapeutic approaches such as check point inhibitors, T-cell therapies, or vaccines.


Assuntos
Azepinas/farmacologia , Células Dendríticas/efeitos dos fármacos , Imunidade/efeitos dos fármacos , Linfocinas/imunologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Receptores de Superfície Celular/antagonistas & inibidores , Linfócitos T/efeitos dos fármacos , Triazóis/farmacologia , Animais , Antígenos/imunologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Citocinas/imunologia , Células Dendríticas/imunologia , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/imunologia , Feminino , Inibidores de Checkpoint Imunológico/farmacologia , Imunidade/imunologia , Imunoterapia/métodos , Contagem de Leucócitos/métodos , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Linfócitos T/imunologia
4.
BMC Cancer ; 17(1): 249, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28388884

RESUMO

BACKGROUND: Epithelial-to-mesenchymal transition (EMT) and cancer stem cells (CSC) contribute to tumour progression and metastasis. Assessment of transcription factors involved in these two mechanisms can help to identify new targets for an oncological therapy. In this study, we focused on the evaluation of the transcription factor Six1 (Sine oculis 1). This protein is involved in embryologic development and its contribution to carcinogenesis has been described in several studies. METHODS: Immunohistochemistry against Six1 was performed on a tissue microarray containing specimens of primary pancreatic ductal adenocarcinomas (PDAC) of 139 patients. Nuclear and cytoplasmic expression was evaluated and correlated to histopathological parameters. Expression of Six1 was inhibited transiently by siRNA in Panc1 and BxPc3 cells and stably by shRNA in Panc1 cells. Expression analysis of CDH1 and Vimentin mRNA was performed and cell motility was tested in a migration assay. Panc1 cells transfected with Six1 shRNA or scrambled shRNA were injected subcutaneously into nude mice. Tumour growth was observed for four weeks. Afterwards, tumours were stained against Six1, CD24 and CD44. RESULTS: Six1 was overexpressed in the cytoplasm and cellular nuclei in malignant tissues (p < 0.0001). No correlation to histopathological parameters could be detected. Six1 down-regulation decreased pancreatic cancer cell motility in vitro. CDH1 and vimentin expression was decreased after inhibition of the expression of Six1. Pancreatic tumours with impaired expression of Six1 showed significantly delayed growth and displayed loss of the CD24+/CD44+ phenotype. CONCLUSION: We show that Six1 is overexpressed in human PDAC and that its inhibition results in a decreased tumour progression in vitro and in vivo. Therefore, targeting Six1 might be a novel therapeutic approach in patients with pancreatic cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Células-Tronco Neoplásicas/metabolismo , Neoplasias Pancreáticas/metabolismo , Interferência de RNA , Animais , Transição Epitelial-Mesenquimal , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Pancreáticas
5.
J Nanobiotechnology ; 14(1): 57, 2016 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-27401816

RESUMO

BACKGROUND: Pancreatic cancer is one of the most lethal of human malignancies known to date and shows relative insensitivity towards most of the clinically available therapy regimens. 3,5-bis(2-fluorobenzylidene)-4-piperidone (EF24), a novel synthetic curcumin analog, has shown promising in vitro therapeutic efficacy in various human cancer cells, but insufficient water solubility and systemic bioavailability limit its clinical application. Here, we describe nano-encapsulation of EF24 into pegylated liposomes (Lipo-EF24) and evaluation of these particles in preclinical in vitro and in vivo model systems of pancreatic cancer. RESULTS: Transmission electron microscopy and size distribution studies by dynamic light scattering confirmed intact spherical morphology of the formed liposomes with an average diameter of less than 150 nm. In vitro, treatment with Lipo-EF24 induced growth inhibition and apoptosis in MIAPaCa and Pa03C pancreatic cancer cells as assessed by using cell viability and proliferation assays, replating and soft agar clonogenicity assays as well as western blot analyses. Lipo-EF24 potently suppressed NF-kappaB nuclear translocation by inhibiting phosphorylation and subsequent degradation of its inhibitor I-kappa-B-alpha. In vivo, synergistic tumor growth inhibition was observed in MIAPaCa xenografts when Lipo-EF24 was given in combination with the standard-of-care cytotoxic agent gemcitabine. In line with in vitro observations, western blot analysis revealed decreased phosphorylation of I-kappa-B-alpha in excised Lipo-EF24-treated xenograft tumor tissues. CONCLUSION: Due to its promising therapeutic efficacy and favorable toxicity profile Lipo-EF24 might be a promising starting point for development of future combinatorial therapeutic regimens against pancreatic cancer.


Assuntos
Antineoplásicos/farmacologia , Compostos de Benzilideno/farmacologia , Desoxicitidina/análogos & derivados , Regulação Neoplásica da Expressão Gênica , Lipossomos/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Piperidonas/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Compostos de Benzilideno/química , Compostos de Benzilideno/farmacocinética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Desoxicitidina/química , Desoxicitidina/farmacocinética , Desoxicitidina/farmacologia , Progressão da Doença , Composição de Medicamentos , Quimioterapia Combinada , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Injeções Subcutâneas , Lipossomos/química , Camundongos , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , NF-kappa B/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Fosforilação/efeitos dos fármacos , Piperidonas/química , Piperidonas/farmacocinética , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
6.
Cancers (Basel) ; 16(10)2024 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-38792002

RESUMO

Bone marrow fibrosis in myeloproliferative neoplasm (MPN), myelodysplastic syndromes (MDS), MPN/MDS overlap syndromes and acute myeloid leukemia (AML) is associated with poor prognosis and early treatment failure. Myelofibrosis (MF) is accompanied by reprogramming of multipotent bone marrow mesenchymal stromal cells (MSC) into osteoid and fiber-producing stromal cells. We demonstrate NRP2 and osteolineage marker NCAM1 (neural cell adhesion molecule 1) expression within the endosteal niche in normal bone marrow and aberrantly in MPN, MDS MPN/MDS overlap syndromes and AML (n = 99), as assessed by immunohistochemistry. Increased and diffuse expression in mesenchymal stromal cells and osteoblasts correlates with high MF grade in MPN (p < 0.05 for NRP2 and NCAM1). Single cell RNA sequencing (scRNAseq) re-analysis demonstrated NRP2 expression in endothelial cells and partial co-expression of NRP2 and NCAM1 in normal MSC and osteoblasts. Potential ligands included transforming growth factor ß1 (TGFB1) from osteoblasts and megakaryocytes. Murine ThPO and JAK2V617F myelofibrosis models showed co-expression of Nrp2 and Ncam1 in osteolineage cells, while fibrosis-promoting MSC only express Nrp2. In vitro experiments with MC3T3-E1 pre-osteoblasts and analysis of Nrp2-/- mouse femurs suggest that Nrp2 is functionally involved in osteogenesis. In summary, NRP2 represents a potential novel druggable target in patients with myelofibrosis.

7.
Hematology ; 28(1): 2212536, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37204122

RESUMO

PURPOSE: Therapeutic regimens and outcome of acute myeloid leukaemia (AML) patients substantially improved over the past decades. However, AML in older patients is still widely understudied and therapeutic standards are far less well defined. This study provides a retrospective analysis of a cohort of AML patients above 65 years of age treated at a single university centre in Germany. METHODS: Treatment regimens including intensive chemotherapy with or without subsequent allogenic stem cell transplantation (allo-SCT), hypomethylating agent (HMA) or low-dose cytarabine (LD-AraC) based therapy or best supportive care (BSC) were evaluated and compared to patient-specific variables, comorbidities indices such as Haematopoietic Cell Transplantation-specific Comorbidity Index (HCT-CI) or Charlson Comorbidity Index (CCI), or Eastern Cooperative Oncology Group (ECOG) performance status to assess their potential impact on outcome. RESULTS: 229 patients ≥ 65 years with newly diagnosed AML were included in this study. Patients received either intensive chemotherapy (IT) without (n = 101, 44%), or followed by allo-SCT (n = 27, 12%), HMA (n = 29, 13%), LD-Ara-C (n = 16, 7%) or best supportive care (BSC) only (n = 56, 24%). Of interest, ECOG performance status predicted overall survival in patients treated with IT, and combinatorial assessment of ECOG and HCT-CI was particularly useful to predict outcome in this subgroup of patients. CONCLUSION: Subsets of AML patients above 65 years of age benefit from intensive chemotherapy and allogenic stem cell transplantation. Combined assessment of ECOG scores and HCT-CI might help to objectively identify suitable patients, and this concept should be further investigated in a prospective manner in future studies.


Selected subsets of AML patients may profit from intensive chemotherapy and allogenic stem cell transplantation.Combined analysis of ECOG performance status and HCT-CI might help to predict outcome in elderly AML patients.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Leucemia Mieloide Aguda , Humanos , Idoso , Estudos Retrospectivos , Resultado do Tratamento , Estudos Prospectivos , Leucemia Mieloide Aguda/tratamento farmacológico , Citarabina , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico
8.
Carcinogenesis ; 33(11): 2242-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22831956

RESUMO

Multiple lines of evidence support a role for curcumin in cancer chemoprevention. Nonetheless, despite its reported efficacy and safety profile, clinical translation of curcumin has been hampered by low oral bioavailability, requiring infeasible 'mega' doses for achieving detectable tissue levels. We have engineered a polymeric nanoparticle encapsulated formulation of curcumin (NanoCurc) to harness its full therapeutic potential. In the current study, we assessed the chemoprevention efficacy of NanoCurc administered via direct intraductal (i.duc) injection in a chemical carcinogen-induced rodent mammary cancer model. Specifically, Sprague-Dawley rats exposed to systemic N-methyl-N-nitrosourea were randomized to receive either oral free curcumin at a previously reported 'mega' dose (200mg/kg) or by direct i.duc injection of free curcumin or NanoCurc, respectively, each delivering 168 µg equivalent of curcumin per rodent teat (a ~20-fold lower dose per animal compared to oral administration). All three chemoprevention modalities resulted in significantly lower mammary tumor incidence compared with control rats; however, there was no significant difference in cancer incidence between the oral dosing and either i.duc arms. On the other hand, mean tumor size, was significantly smaller in the i.duc NanoCurc cohort compared with i.duc free curcumin (P < 0.0001), suggesting the possibility of better resectability for 'breakthrough' cancers. Reduction in cancer incidence was associated with significant decrease in nuclear factor -κB activation in the NanoCurc treated mammary epithelium explants, compared to either control or oral curcumin-administered rats. Our studies confirm the potential for i.duc NanoCurc as an alternative to the oral route for breast cancer chemoprevention in high-risk cohorts.


Assuntos
Antineoplásicos/administração & dosagem , Curcumina/administração & dosagem , Modelos Animais de Doenças , Neoplasias Mamárias Experimentais/prevenção & controle , Nanopartículas , Polímeros/química , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Disponibilidade Biológica , Química Farmacêutica , Curcumina/farmacocinética , Curcumina/farmacologia , Relação Dose-Resposta a Droga , Feminino , Técnicas Imunoenzimáticas , Incidência , Neoplasias Mamárias Experimentais/epidemiologia , Neoplasias Mamárias Experimentais/mortalidade , Metilnitrosoureia/toxicidade , Ratos , Ratos Sprague-Dawley , Taxa de Sobrevida , Distribuição Tecidual
9.
Lab Invest ; 91(9): 1383-95, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21691262

RESUMO

Plant-derived polyphenols such as curcumin hold promise as a therapeutic agent in the treatment of chronic liver diseases. However, its development is plagued by poor aqueous solubility resulting in poor bioavailability. To circumvent the suboptimal bioavailability of free curcumin, we have developed a polymeric nanoparticle formulation of curcumin (NanoCurc™) that overcomes this major pitfall of the free compound. In this study, we show that NanoCurc™ results in sustained intrahepatic curcumin levels that can be found in both hepatocytes and non-parenchymal cells. NanoCurc™ markedly inhibits carbon tetrachloride-induced liver injury, production of pro-inflammatory cytokines and fibrosis. It also enhances antioxidant levels in the liver and inhibits pro-fibrogenic transcripts associated with activated myofibroblasts. Finally, we show that NanoCurc™ directly induces stellate cell apoptosis in vitro. Our results suggest that NanoCurc™ might be an effective therapy for patients with chronic liver disease.


Assuntos
Intoxicação por Tetracloreto de Carbono/prevenção & controle , Curcumina/química , Mediadores da Inflamação/antagonistas & inibidores , Nanopartículas , Animais , Disponibilidade Biológica , Intoxicação por Tetracloreto de Carbono/metabolismo , Linhagem Celular Transformada , Curcumina/farmacocinética , Curcumina/uso terapêutico , Curcumina/toxicidade , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Oncol Res Treat ; 44(10): 538-546, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34515189

RESUMO

OBJECTIVE: We report the spectrum of immune-related adverse effects (irAEs) and outcome observed in a single-center cohort of 100 patients treated with immune checkpoint inhibitors in a routine clinical setting. METHODS: Tumor entities included non-small cell lung cancer (n = 28), head and neck squamous cell carcinoma (n = 28), urothelial carcinoma (n = 7), and others (n = 37). RESULTS: irAEs were documented in 49% of cases analyzed, and the most frequent manifestation consisted of immune-mediated skin rash (28%), colitis (9%), pneumonitis (8%), hypothyroidism (7%), or hepatitis (6%). Skin rash correlated with improved progression-free survival. CONCLUSION: Development of immune-related skin rash was found to correlate with favorable outcome, suggesting its practical feasibility as a potential predictive surrogate marker.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Carcinoma de Células de Transição , Exantema , Neoplasias de Cabeça e Pescoço , Neoplasias Pulmonares , Neoplasias da Bexiga Urinária , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Exantema/induzido quimicamente , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Humanos , Inibidores de Checkpoint Imunológico , Imunoterapia/efeitos adversos , Neoplasias Pulmonares/tratamento farmacológico , Receptor de Morte Celular Programada 1 , Carcinoma de Células Escamosas de Cabeça e Pescoço
11.
Int J Endocrinol ; 2020: 1491475, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32089680

RESUMO

OBJECTIVE: Neuroendocrine neoplasias (NENs) represent a rare and biologically heterogeneous group of malignancies. Treatment of NEN patients remains challenging due to lack of prospective evidence on the choice of ideal therapeutic sequence and therapeutic efficacy in specific individual scenarios. METHODS: Clinical data on 110 consecutive patients suffering from NEN treated at a single German university center were analyzed, therapeutic regimens applied were assessed, and the outcome was evaluated. RESULTS: Histological grading, Ki67 proliferation index, functional activity, and presence of metastases were identified as prognostic markers. 10-year overall survival rates were 92%, 44%, and 0% for G1, G2, and G3 tumors, and 60%, 39%, 69%, 53%, and 0% for Ki67 <2%, 3-5%, 6-20%, 21-49%, and >50%, respectively. Peptide receptor radionuclide therapy (PRRT) and cytostatic chemotherapy were the second most common options, with PRRT being used more frequently in NET G1 and G2 and chemotherapy in NEC G3. Combination chemotherapy with etoposide plus cisplatin or carboplatin showed disease control rates (DCRs) of overall 74%, with a short median progression-free survival (PFS) of 7 or 5 months, respectively. DCR and PFS for PRRT were 89% and 22 months when administered as monotherapy, versus 100% and 27 months upon combination with somatostatin analog (SSA) therapy. Of note, PRRT also achieved disease control as best response in 5/5 (100%) selected cases of NEC G3. CONCLUSION: Further prospective studies are warranted to help stratify available options for therapeutic intervention in NEN patients.

12.
Mol Cancer Ther ; 7(12): 3878-88, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19074860

RESUMO

Nanotechnology has enabled significant advances in the areas of cancer diagnosis and therapy. The field of drug delivery is a sterling example, with nanoparticles being increasingly used for generating therapeutic formulations of poorly water-soluble, yet potent anticancer drugs. Whereas a number of nanoparticle-drug combinations are at various stages of preclinical or clinical assessment, the overwhelming majorities of such systems are injectable formulations and are incapable of being partaken orally. The development of an oral nano-delivery system would have distinct advantages for cancer chemotherapy. We report the synthesis and physicochemical characterization of orally bioavailable polymeric nanoparticles composed of N-isopropylacrylamide, methylmethacrylate, and acrylic acid in the molar ratios of 60:20:20 (designated NMA622). Amphiphilic NMA622 nanoparticles show a size distribution of <100 nm (mean diameter of 80 +/- 34 nm) with low polydispersity and can readily encapsulate a number of poorly water-soluble drugs such as rapamycin within the hydrophobic core. No apparent systemic toxicities are observed in mice receiving as much as 500 mg/kg of the orally administered void NMA622 for 4 weeks. Using NMA622-encapsulated rapamycin ("nanorapamycin") as a prototype for oral nano-drug delivery, we show favorable in vivo pharmacokinetics and therapeutic efficacy in a xenograft model of human pancreatic cancer. Oral nanorapamycin leads to robust inhibition of the mammalian target of rapamycin pathway in pancreatic cancer xenografts, which is accompanied by significant growth inhibition (P < 0.01) compared with control tumors. These data indicate that NMA622 nanoparticles provide a suitable platform for oral delivery of water-insoluble drugs like rapamycin for cancer therapy.


Assuntos
Sistemas de Liberação de Medicamentos , Nanopartículas/química , Polímeros/química , Administração Oral , Animais , Linhagem Celular Tumoral , Química Farmacêutica/métodos , Desenho de Fármacos , Humanos , Cinética , Camundongos , Transplante de Neoplasias , Neoplasias Pancreáticas/tratamento farmacológico , Sirolimo/farmacologia , Tecnologia Farmacêutica/métodos
13.
Mol Cancer Ther ; 7(9): 2725-35, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18790753

RESUMO

Recent evidence suggests that blockade of aberrant Hedgehog signaling can be exploited as a therapeutic strategy for pancreatic cancer. Our previous studies using the prototype Hedgehog small-molecule antagonist cyclopamine had shown the striking inhibition of systemic metastases on Hedgehog blockade in spontaneously metastatic orthotopic xenograft models. Cyclopamine is a natural compound with suboptimal pharmacokinetics, which impedes clinical translation. In the present study, a novel, orally bioavailable small-molecule Hedgehog inhibitor, IPI-269609, was tested using in vitro and in vivo model systems. In vitro treatment of pancreatic cancer cell lines with IPI-269609 resembled effects observed using cyclopamine (i.e., Gli-responsive reporter knockdown, down-regulation of the Hedgehog target genes Gli1 and Ptch, as well as abrogation of cell migration and colony formation in soft agar). Single-agent IPI-269609 profoundly inhibited systemic metastases in orthotopic xenografts established from human pancreatic cancer cell lines, although Hedgehog blockade had minimal effect on primary tumor volume. The only discernible phenotype observed within the treated primary tumor was a significant reduction in the population of aldehyde dehydrogenase-bright cells, which we have previously identified as a clonogenic tumor-initiating population in pancreatic cancer. Selective ex vivo depletion of aldehyde dehydrogenase-bright cells with IPI-269609 was accompanied by significant reduction in tumor engraftment rates in athymic mice. Pharmacologic blockade of aberrant Hedgehog signaling might prove to be an effective therapeutic strategy for inhibition of systemic metastases in pancreatic cancer, likely through targeting subsets of cancer cells with tumor-initiating ("cancer stem cell") properties.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/farmacocinética , Azulenos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Proteínas Hedgehog/antagonistas & inibidores , Neoplasias Pancreáticas/patologia , Transdução de Sinais/efeitos dos fármacos , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Azulenos/administração & dosagem , Azulenos/química , Disponibilidade Biológica , Compostos Bicíclicos Heterocíclicos com Pontes/administração & dosagem , Compostos Bicíclicos Heterocíclicos com Pontes/química , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Nus , Metástase Neoplásica , Neoplasias Pancreáticas/tratamento farmacológico , Receptores Patched , Receptor Patched-1 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Ensaio Tumoral de Célula-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Expert Opin Drug Discov ; 14(2): 127-142, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30657339

RESUMO

INTRODUCTION: Despite the introduction of novel therapeutic regimens for advanced stages of pancreatic cancer, long-term survival and overall outcome for patients are still very poor. Suitable small animal models are a prerequisite for better understanding of underlying pathophysiology and for translational studies designed to uncover novel therapeutic targets and to evaluate therapy regimens on a preclinical level. Areas covered: Genetically engineered mouse models as well as syngenic and xenotransplantation models are summarized and critically discussed with respect to their value for pancreatic cancer translational research. Expert opinion: Mouse models of pancreatic cancer represent a valuable tool for translational studies and are indispensable in order to develop novel potent drugs against pancreatic cancer. More complex genetically engineered mouse models are being developed to cover the complex genomic alterations found in human pancreatic cancers and to enable studies correlating genotype with response to specific therapeutic interventions. Another promising area of research in this field is the refinement and further development of humanized patient-derived xenograft models.


Assuntos
Antineoplásicos/farmacologia , Descoberta de Drogas/métodos , Neoplasias Pancreáticas/tratamento farmacológico , Animais , Modelos Animais de Doenças , Engenharia Genética , Humanos , Camundongos , Neoplasias Pancreáticas/patologia , Pesquisa Translacional Biomédica/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Oncol ; 2019: 8701824, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30949204

RESUMO

Therapeutic options for advanced stage cholangiocellular carcinoma (CCC) are very limited as of today and patients carry an exceptionally poor overall prognosis. In recent years, increasing evidence has been accumulated to suggest that malignant cells widely show increased intrinsic ROS levels and exhibit altered redox profiles as compared to normal counterparts, opening up potential avenues for therapeutic intervention. This study provides preclinical experimental evidence of therapeutic activity of the curcumin analog EF24 in cholangiocarcinoma models. In CCC cell lines, EF24 inhibited cell viability and induced apoptosis through excessive ROS generation. Moreover, administration of EF24 led to depletion of total intracellular GSH levels, induced mitochondrial depolarization, and abrogated STAT3 phosphorylation. Of interest, these effects were readily averted by treating the cells with exogenous antioxidants such as N-acetyl cysteine (NAC) or glutathione monoethyl ester (GEE). In vivo, EF24, solubilized using a cyclodextrin formulation, significantly suppressed the growth of tumor xenografts without exhibiting any toxic adverse effects. Immunohistochemical analysis of extracted tumor tissues demonstrated reduced nuclear staining for Ki-67 and downregulation of phospho-STAT3 as well as strong staining for oxidative stress biomarker 8-OHdG. Therefore, the data presented here suggest EF24 as potential therapeutic compound against CCC which might act at least to some extent through ROS-induced oxidative damage, subsequently inducing apoptosis. Further evaluation of this approach should be carried out in future follow-up studies.

16.
Oncol Res Treat ; 41(10): 596-602, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30269126

RESUMO

Pancreatic ductal adenocarcinoma (PDAC, pancreatic cancer) carries one of the poorest overall prognoses of all human malignancies known to date. Despite the introduction of novel therapeutic regimens, the outcome has not markedly improved over the past decades, the incidence rates are almost identical to the mortality rates, and PDAC is projected to soon become the second most common cause of cancer-related mortality in Western countries. Despite this clear medical need to develop novel therapeutic strategies against this dire malady, this need has so far not been addressed with sufficient institutional attention and support in terms of research funding and strategical programs. Given the still growing life expectancy and projected demographic changes with a growing proportion of senior citizens in many European societies, this discrepancy is likely to become even more pressing in the future. This article provides a brief overview of ongoing preclinical efforts to identify novel targets and, based on this, to develop novel strategies to treat advanced pancreatic cancer and improve survival and the quality of life of patients suffering from this malignancy.


Assuntos
Neoplasias Pancreáticas/terapia , Quinase 5 Dependente de Ciclina/antagonistas & inibidores , Dano ao DNA , Galectina 3/antagonistas & inibidores , Humanos , Imunoterapia , Biópsia Líquida , Mutação , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Transdução de Sinais/fisiologia
17.
Oncol Res Treat ; 41(10): 590-594, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30286472

RESUMO

Pancreatic cancer remains the fourth most common cause of cancer-related mortality and is a major health threat. The majority of cases are diagnosed at advanced disease stages, limiting the chances of long-term survival. Several new therapeutic regimens have been introduced into routine clinical practice in recent years and a plethora of novel approaches is currently undergoing preclinical and early clinical evaluation. This review discusses the current standards of care for systemic therapy of pancreatic cancer and gives a brief outlook on ongoing clinical trials.


Assuntos
Carcinoma Ductal Pancreático/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Ductal Pancreático/patologia , Ensaios Clínicos como Assunto , Humanos , Cuidados Paliativos , Neoplasias Pancreáticas/patologia
18.
Leukemia ; 32(11): 2445-2458, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29654274

RESUMO

Macrophages are key mediators of the therapeutic effects exerted by monoclonal antibodies, such as the anti-CD38 antibody MOR202, currently introduced in multiple myeloma (MM) therapy. Therefore, it is important to understand how antibody-mediated effector functions of myeloma-associated macrophages (MAMs) are regulated. Here, we focused on the effects of vitamin D, a known regulator of macrophage effector functions. Consequently, it was the aim of this study to assess whether modulation of the vitamin D pathway alters the tumoricidal activity of MAMs. Here, we demonstrate that MAMs display a defective vitamin D pathway with reduced expression level of CYP27B1 and limited tumoricidal activity which can be restored by the IMiD lenalidomide in vitro. Furthermore, our data indicate that the vitamin D pathway of MAMs from MM patients does recover during an IMiD-containing therapy shown by an improved MOR202-mediated cytotoxic activity of these MAMs against primary MM cells ex vivo. Here, the ex vivo cytotoxic activity could be further enhanced by vitamin D supplementation. These data suggest that vitamin D holds a key role for the effector functions of MAMs and that vitamin D supplementation in IMiD combination trials could further increase the therapeutic efficacy of anti-CD38 antibodies such as MOR202, which remains to be investigated in clinical studies.


Assuntos
Anticorpos Monoclonais/farmacologia , Lenalidomida/farmacologia , Macrófagos/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Vitamina D/metabolismo , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/metabolismo , ADP-Ribosil Ciclase 1/metabolismo , Linhagem Celular Tumoral , Citotoxinas/farmacologia , Humanos , Macrófagos/metabolismo
19.
J Nanobiotechnology ; 5: 3, 2007 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-17439648

RESUMO

BACKGROUND: Curcumin, a yellow polyphenol extracted from the rhizome of turmeric (Curcuma longa), has potent anti-cancer properties as demonstrated in a plethora of human cancer cell line and animal carcinogenesis models. Nevertheless, widespread clinical application of this relatively efficacious agent in cancer and other diseases has been limited due to poor aqueous solubility, and consequently, minimal systemic bioavailability. Nanoparticle-based drug delivery approaches have the potential for rendering hydrophobic agents like curcumin dispersible in aqueous media, thus circumventing the pitfalls of poor solubility. RESULTS: We have synthesized polymeric nanoparticle encapsulated formulation of curcumin - nanocurcumin - utilizing the micellar aggregates of cross-linked and random copolymers of N-isopropylacrylamide (NIPAAM), with N-vinyl-2-pyrrolidone (VP) and poly(ethyleneglycol)monoacrylate (PEG-A). Physico-chemical characterization of the polymeric nanoparticles by dynamic laser light scattering and transmission electron microscopy confirms a narrow size distribution in the 50 nm range. Nanocurcumin, unlike free curcumin, is readily dispersed in aqueous media. Nanocurcumin demonstrates comparable in vitro therapeutic efficacy to free curcumin against a panel of human pancreatic cancer cell lines, as assessed by cell viability and clonogenicity assays in soft agar. Further, nanocurcumin's mechanisms of action on pancreatic cancer cells mirror that of free curcumin, including induction of cellular apoptosis, blockade of nuclear factor kappa B (NFkappaB) activation, and downregulation of steady state levels of multiple pro-inflammatory cytokines (IL-6, IL-8, and TNFalpha). CONCLUSION: Nanocurcumin provides an opportunity to expand the clinical repertoire of this efficacious agent by enabling ready aqueous dispersion. Future studies utilizing nanocurcumin are warranted in pre-clinical in vivo models of cancer and other diseases that might benefit from the effects of curcumin.

20.
Int J Pharm ; 288(1): 157-68, 2005 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-15607268

RESUMO

Nanoparticles of calcium phosphate encapsulating plasmid DNA (pDNA) of size 100-120 nm in diameter were prepared. XRD studies of these nanoparticles showed them to be crystalline in nature having hydroxyapatite structure. The maximum loading of pDNA and its release from nanoparticles were studied using gel electrophoresis. The time dependent size measurement of these particles demonstrated that these particles show strong aggregational behaviour in aqueous dispersion. Calcium phosphate nanoparticles were found to be dissolved even in low acidic buffer (pH 5.0) releasing the pDNA, which suggested that DNA release from these particles in the endosomal compartment was possible. In vitro transfection efficiency of these calcium phosphate nanoparticles was found to be higher than that of the commercial transfecting reagent Polyfect.


Assuntos
Fosfatos de Cálcio/administração & dosagem , Vetores Genéticos/administração & dosagem , Nanoestruturas , Plasmídeos/administração & dosagem , DNA/administração & dosagem , DNA/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Plasmídeos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA