Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 7(41): 67166-67174, 2016 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-27564103

RESUMO

Fc gamma receptor I (FcγRI, CD64) is a well-known target antigen for passive immunotherapy against acute myeloid leukemia and chronic myelomonocytic leukemia. We recently reported the preclinical immunotherapeutic potential of microtubule associated protein tau (MAP) against a variety of cancer types including breast carcinoma and Hodgkin's lymphoma. Here we demonstrate that the CD64-directed human cytolytic fusion protein H22(scFv)-MAP kills ex vivo 15-50% of CD64+ leukemic blasts derived from seven myeloid leukemia patients. Furthermore, in contrast to the nonspecific cytostatic agent paclitaxel, H22(scFv)-MAP showed no cytotoxicity towards healthy CD64+ PBMC-derived cells and macrophages. The targeted delivery of this microtubule stabilizing agent therefore offers a promising new strategy for specific treatment of CD64+ leukemia.


Assuntos
Antineoplásicos/farmacologia , Leucemia Mieloide Aguda , Proteínas Associadas aos Microtúbulos/farmacologia , Terapia de Alvo Molecular/métodos , Receptores de IgG , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Feminino , Humanos , Imunotoxinas/farmacologia , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes de Fusão/farmacologia
2.
Oncotarget ; 7(34): 54925-54936, 2016 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-27448975

RESUMO

Triple-negative breast cancer (TNBC) is a heterogeneous disease in which the tumors do not express estrogen receptor (ER), progesterone receptor (PgR) or human epidermal growth factor receptor 2 (HER2). Classical receptor-targeted therapies such as tamoxifen or trastuzumab are therefore unsuitable and combinations of surgery, chemotherapy and/or radiotherapy are required. Photoimmunotheranostics is a minimally invasive approach in which antibodies deliver nontoxic photosensitizers that emit light to facilitate diagnosis and produce cytotoxic reactive oxygen species to induce apoptosis and/or necrosis in cancer cells. We developed a panel of photoimmunotheranostic agents against three TNBC-associated cell surface antigens. Antibodies against epidermal growth factor receptor (EGFR), epithelial cell adhesion molecule (EpCAM) and chondroitin sulfate proteoglycan 4 (CSPG4) were conjugated to the highly potent near-infrared imaging agent/photosensitizer IRDye®700DX phthalocyanine using SNAP-tag technology achieving clear imaging in both breast cancer cell lines and human biopsies and highly potent phototherapeutic activity with IC50values of 62-165 nM against five different cell lines expressing different levels of EGFR, EpCAM and CSPG4. A combination of all three reagents increased the therapeutic activity against TNBC cells by up to 40%.


Assuntos
Imunoconjugados/uso terapêutico , Indóis/uso terapêutico , Compostos de Organossilício/uso terapêutico , Fotoquimioterapia/métodos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Proteoglicanas de Sulfatos de Condroitina/imunologia , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Molécula de Adesão da Célula Epitelial/imunologia , Molécula de Adesão da Célula Epitelial/metabolismo , Receptores ErbB/imunologia , Receptores ErbB/metabolismo , Feminino , Guanina/análogos & derivados , Guanina/química , Humanos , Imunoconjugados/química , Indóis/química , Isoindóis , Luz , Células MCF-7 , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Microscopia Confocal , Compostos de Organossilício/química , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/uso terapêutico , Proteínas Recombinantes de Fusão/química , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/imunologia , Neoplasias de Mama Triplo Negativas/diagnóstico , Neoplasias de Mama Triplo Negativas/metabolismo
3.
Int J Cancer ; 139(4): 916-27, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27037627

RESUMO

Chondroitin sulfate proteoglycan 4 (CSPG4) has been identified as a highly promising target antigen for immunotherapy of triple-negative breast cancer (TNBC). TNBC represents a highly aggressive heterogeneous group of tumors lacking expression of estrogen, progesterone and human epidermal growth factor receptor 2. TNBC is particularly prevalent among young premenopausal women. No suitable targeted therapies are currently available and therefore, novel agents for the targeted elimination of TNBC are urgently needed. Here, we present a novel cytolytic fusion protein (CFP), designated αCSPG4(scFv)-MAP, that consists of a high affinity CSPG4-specific single-chain antibody fragment (scFv) genetically fused to a functionally enhanced form of the human microtubule-associated protein (MAP) tau. Our data indicate that αCSPG4(scFv)-MAP efficiently targets CSPG4(+) TNBC-derived cell lines MDA-MB-231 and Hs 578T and potently inhibits their growth with IC50 values of ∼200 nM. Treatment with αCSPG(scFv)-MAP resulted in induction of the mitochondrial stress pathway by activation of caspase-9 as well as endonuclease G translocation to the nucleus, while induction of the caspase-3 apoptosis pathway was not detectable. Importantly, in vivo studies in mice bearing human breast cancer xenografts revealed efficient targeting to and accumulation of αCSPG4(scFv)-MAP at tumor sites resulting in prominent tumor regression. Taken together, this preclinical proof of concept study confirms the potential clinical value of αCSPG4(scFv)-MAP as a novel targeted approach for the elimination of CSPG4-positive TNBC.


Assuntos
Anticorpos Monoclonais/genética , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Anticorpos de Cadeia Única/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Proteínas tau/metabolismo , Animais , Biomarcadores , Biomarcadores Tumorais , Caspase 3/metabolismo , Caspase 9/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Proteoglicanas de Sulfatos de Condroitina/genética , Modelos Animais de Doenças , Feminino , Expressão Gênica , Humanos , Proteínas de Membrana/genética , Camundongos , Terapia de Alvo Molecular , Ligação Proteica , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Tubulina (Proteína)/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas tau/genética
4.
Cancer Lett ; 372(2): 201-9, 2016 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-26806809

RESUMO

Triple-negative breast cancer (TNBC) is associated with poor prognosis and high prevalence among young premenopausal women. Unlike in other breast cancer subtypes, no targeted therapy is currently available. Overexpression of epithelial cell adhesion molecule (EpCAM) in 60% of TNBC tumors correlates with poorer prognosis and is associated with cancer stem cell phenotype. Thus, selective elimination of EpCAM(+) TNBC tumor cells is of clinical importance. Therefore, we constructed a fully human targeted cytolytic fusion protein, designated GbR201K-αEpCAM(scFv), in which an EpCAM-selective single-chain antibody fragment (scFv) is genetically fused to a granzyme B (Gb) mutant with reduced sensitivity to its natural inhibitor serpin B9. In vitro studies confirmed its specific binding, internalization and cytotoxicity toward a panel of EpCAM-expressing TNBC cells. Biodistribution kinetics and tumor-targeting efficacy using MDA-MB-468 cells in a human TNBC xenograft model in mice revealed selective accumulation of GbR201K-αEpCAM(scFv) in the tumors after i.v. injection. Moreover, treatment of tumor-bearing mice demonstrated a prominent inhibition of tumor growth of up to 50 % in this proof-of-concept study. Taken together, our results indicate that GbR201K-αEpCAM(scFv) is a promising novel targeted therapeutic for the treatment of TNBC.


Assuntos
Apoptose/efeitos dos fármacos , Moléculas de Adesão Celular/antagonistas & inibidores , Granzimas/farmacologia , Imunoterapia/métodos , Imunotoxinas/farmacologia , Anticorpos de Cadeia Única/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Carga Tumoral/efeitos dos fármacos , Animais , Especificidade de Anticorpos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Molécula de Adesão da Célula Epitelial , Feminino , Granzimas/genética , Granzimas/farmacocinética , Células HEK293 , Humanos , Imunotoxinas/genética , Imunotoxinas/imunologia , Imunotoxinas/farmacocinética , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação , Proteínas Recombinantes de Fusão/farmacologia , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/farmacocinética , Distribuição Tecidual , Transfecção , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Immunol ; 194(11): 5253-60, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25917102

RESUMO

The hormone leptin plays a key role in energy homeostasis, and the absence of either leptin or its receptor (LepR) leads to severe obesity and metabolic disorders. To avoid indirect effects and to address the cell-intrinsic role of leptin signaling in the immune system, we conditionally targeted LepR in T cells. In contrast with pleiotropic immune disorders reported in obese mice with leptin or LepR deficiency, we found that LepR deficiency in CD4(+) T cells resulted in a selective defect in both autoimmune and protective Th17 responses. Reduced capacity for differentiation toward a Th17 phenotype by lepr-deficient T cells was attributed to reduced activation of the STAT3 and its downstream targets. This study establishes cell-intrinsic roles for LepR signaling in the immune system and suggests that leptin signaling during T cell differentiation plays a crucial role in T cell peripheral effector function.


Assuntos
Diferenciação Celular/imunologia , Leptina/imunologia , Obesidade/imunologia , Receptores para Leptina/imunologia , Células Th17/citologia , Animais , Autoimunidade/genética , Autoimunidade/imunologia , Diferenciação Celular/genética , Células Cultivadas , Citrobacter rodentium/imunologia , Colite/imunologia , Infecções por Enterobacteriaceae/imunologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/genética , Receptores para Leptina/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Células Th17/imunologia
6.
Mol Cancer Ther ; 13(9): 2194-202, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24980949

RESUMO

In normal epithelia, the epithelial cell adhesion molecule (EpCAM) expression is relatively low and only present at the basolateral cell surface. In contrast, EpCAM is aberrantly overexpressed in various human carcinomas. Therefore, EpCAM is considered to be a highly promising target for antibody-based cancer immunotherapy. Here, we present a new and fully human cytolytic fusion protein (CFP), designated "anti-EpCAM(scFv)-MAP," that is comprised of an EpCAM-specific antibody fragment (scFv) genetically fused to the microtubule-associated protein tau (MAP). Anti-EpCAM(scFv)-MAP shows potent EpCAM-restricted proapoptotic activity toward rapidly proliferating carcinoma cells. In vitro assays confirmed that treatment with anti-EpCAM(scFv)-MAP resulted in the colocalization and stabilization of microtubules, suggesting that this could be the potential mode of action. Dose-finding experiments indicated that anti-EpCAM(scFv)-MAP is well tolerated in mice. Using noninvasive far-red in vivo imaging in a tumor xenograft mouse model, we further demonstrated that anti-EpCAM(scFv)-MAP inhibited tumor growth in vivo. In conclusion, our data suggest that anti-EpCAM(scFv)-MAP may be of therapeutic value for the targeted elimination of EpCAM(+) carcinomas.


Assuntos
Antígenos de Neoplasias/química , Moléculas de Adesão Celular/química , Neoplasias/terapia , Proteínas tau/química , Animais , Apoptose , Carcinoma/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Molécula de Adesão da Célula Epitelial , Feminino , Células HEK293 , Humanos , Imunoterapia/métodos , Concentração Inibidora 50 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Fases de Leitura Aberta , Ligação Proteica , Proteínas Recombinantes de Fusão/química , Tubulina (Proteína)/química
7.
Curr Pharm Des ; 19(30): 5429-36, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23431985

RESUMO

Although current cancer treatment strategies are highly aggressive, they are often not effective enough to destroy the collectivity of malignant cells. The residual tumor cells that survived the first-line treatment may continue to proliferate or even metastasize. Therefore, the development of novel more effective strategies to specifically eliminate also single cancer cells is urgently needed. In this respect, the development of antibody-based therapeutics, in particular example immunotoxins, has attracted broad interest. Since the internalization of immunotoxins is essential for their cytotoxic effectivity, it is of crucial importance to study their internalization behavior to assess the potential for their therapeutic use. In this study, we determined the internalization behavior of four different single-chain fragments variable (scFv) when binding to the corresponding target antigen as expressed on solid or non-solid tumor cell lines. The scFvs were recombinantly fused to the SNAP-tag, an engineered variant of the human repair enzyme O(6)-alkylguanine-DNA alkyltransferase that covalently reacts with benzylguanine derivatives. Since a large number of highly sensitive organic fluorescent dyes are already available or can easily be derivatized to react with the self-labeling SNAP-tag, this system provides versatile applications for imaging of intraand extracellular compartments of living cells. The fusion proteins were coupled to SNAP-surface(®) Alexa Fluor(®) 488 or SNAP-surface(®) Alexa Fluor(®) 647 and binding as well as internalization was monitored by flow cytometry and confocal microscopy, respectively. Depending on the respective target antigen, we could distinguish between slow and rapid internalization behavior. Moreover, we detected increased internalization rate for bivalent scFv constructs. Our approach allows for rapid and early stage evaluation of the internalization characteristics of new antibodies designated for further therapeutic development.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Imagem Molecular/métodos , Neoplasias/metabolismo , Animais , Anticorpos , Clonagem Molecular , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Neoplasias/tratamento farmacológico , Ligação Proteica
8.
Curr Pharm Des ; 19(30): 5437-42, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23431986

RESUMO

In the past two decades, immense advances have been achieved in the engineering, production and purifying of recombinant proteins. These proteins are being widely utilized in many fields of biology, biotechnology and medicine, including diagnostic and therapeutic applications. These applications often require the modification or conjugation of these proteins with other molecules. Researchers are spending many efforts to develop and improve the methods of protein modifications. A main challenge they face is derivatizing proteins without affecting their structure and biological function. The conjugation methods available today include random and specific chemical modifications on endogenous amino acids or carbohydrate of the protein of interest. Other methods utilize self-labeling tags as fusion partners to the original protein enabling site-specific conjugation. SNAP-tag is one of the most promising self-labeling tags, which reacts specifically, rapidly and covalently with benzylguanine (BG) derivatives. SNAP-tag fusion proteins have been successfully used for imaging living cells. Recently, several studies have utilized the SNAP technology for generating antibody-based diagnostic and therapeutic tools. We here review these approaches and their possible impact on improving cancer targeting.


Assuntos
Imagem Molecular/métodos , Proteínas/química , Proteínas/metabolismo , Animais , Anticorpos , Humanos , Sondas Moleculares , Preparações Farmacêuticas , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA