Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Diagn ; 20(3): 344-354, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29471115

RESUMO

Myelodysplastic syndromes are hematological neoplasias in which immunohistologic examination of bone marrow trephines is important for a definite diagnosis. Unequivocal distinction from reactive bone marrow changes is, however, sometimes difficult. Because neoplastic clones in myelodysplastic syndrome carry mutations in recurrent genes, mutation detection by targeted next-generation sequencing may be a useful support for differential diagnosis. To elucidate the accuracy of this approach in the clinical diagnostic setting, we analyzed single and consecutive bone marrow trephines processed for immunohistologic examination from 145 patients by targeted next-generation sequencing of 12 genes recurrently mutated in myelodysplastic syndromes. Of 110 patients with immunohistologic unequivocal diagnosis, 41 of 47 with myelodysplastic syndrome carried mutations. In 14 consecutive samples available from these patients, remissions were accompanied by loss of mutations and ongoing disease with persisting mutations. Of 35 samples with indefinite immunohistologic appearance, 22 developed clinical unequivocal myelodysplastic syndrome in the further course, and 19 carried mutations already in the initial biopsy, which persisted in consecutive samples available from 13 patients. No mutation was detected in any initial and consecutive sample of 13 patients with indefinite immunohistologic appearance without clinical unequivocal myelodysplastic syndrome in the further course. We conclude that targeted next-generation sequencing is an accurate tool for differential diagnosis of myelodysplastic syndrome in the clinical diagnostic setting.


Assuntos
Medula Óssea/patologia , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Síndromes Mielodisplásicas/diagnóstico , Síndromes Mielodisplásicas/genética , Células Clonais , Diagnóstico Diferencial , Humanos , Mutação/genética , Taxa de Mutação , Síndromes Mielodisplásicas/patologia
2.
J Cancer Res Clin Oncol ; 143(10): 2025-2038, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28667390

RESUMO

PURPOSE: Despite great progress in the diagnosis and treatment of localized prostate cancer (PCa), there remains a need for new diagnostic markers that can accurately distinguish indolent and aggressive variants. One promising approach is the antibody-based targeting of prostate stem cell antigen (PSCA), which is frequently overexpressed in PCa. Here, we show the construction of a molecular imaging probe comprising a humanized scFv fragment recognizing PSCA genetically fused to an engineered version of the human DNA repair enzyme O6-alkylguanine-DNA alkyltransferase (AGT), the SNAP-tag, enabling specific covalent coupling to various fluorophores for diagnosis of PCa. Furthermore, the recombinant immunotoxin (IT) PSCA(scFv)-ETA' comprising the PSCA(scFv) and a truncated version of Pseudomonas exotoxin A (PE, ETA') was generated. METHODS: We analyzed the specific binding and internalization behavior of the molecular imaging probe PSCA(scFv)-SNAP in vitro by flow cytometry and live cell imaging, compared to the corresponding IT PSCA(scFv)-ETA'. The cytotoxic activity of PSCA(scFv)-ETA' was tested using cell viability assays. Specific binding was confirmed on formalin-fixed paraffin-embedded tissue specimen of early and advanced PCa. RESULTS: Alexa Fluor® 647 labeling of PSCA(scFv)-SNAP confirmed selective binding to PSCA, leading to rapid internalization into the target cells. The recombinant IT PSCA(scFv)-ETA' showed selective binding leading to internalization and efficient elimination of target cells. CONCLUSIONS: Our data demonstrate, for the first time, the specific binding, internalization, and cytotoxicity of a scFv-based fusion protein targeting PSCA. Immunohistochemical staining confirmed the specific ex vivo binding to primary PCa material.


Assuntos
ADP Ribose Transferases/administração & dosagem , Antígenos de Neoplasias/imunologia , Toxinas Bacterianas/administração & dosagem , Exotoxinas/administração & dosagem , Imunotoxinas/administração & dosagem , Proteínas de Neoplasias/imunologia , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/terapia , Proteínas Recombinantes de Fusão/administração & dosagem , Fatores de Virulência/administração & dosagem , ADP Ribose Transferases/imunologia , Toxinas Bacterianas/imunologia , Exotoxinas/imunologia , Citometria de Fluxo , Proteínas Ligadas por GPI/imunologia , Células HEK293 , Humanos , Testes Imunológicos , Imunoterapia/métodos , Imunotoxinas/imunologia , Imunotoxinas/farmacocinética , Masculino , Terapia de Alvo Molecular , Neoplasias da Próstata/imunologia , Proteínas Recombinantes de Fusão/imunologia , Anticorpos de Cadeia Única/administração & dosagem , Anticorpos de Cadeia Única/imunologia , Fatores de Virulência/imunologia , Exotoxina A de Pseudomonas aeruginosa
3.
Cancer Lett ; 365(2): 149-55, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25888452

RESUMO

The treatment of rhabdomyosarcoma (RMS) is challenging, and the prognosis remains especially poor for high-grade RMS with metastasis. The conventional treatment of RMS is based on multi-agent chemotherapy combined with resection and radiotherapy, which are often marked by low success rate. Alternative therapeutic options include the combination of standard treatments with immunotherapy. We generated a microtubule-associated protein (MAP)-based fully human cytolytic fusion protein (hCFP) targeting the fetal acetylcholine receptor, which is expressed on RMS cells. We were able to express and purify functional scFv35-MAP from Escherichia coli cells. Moreover, we found that scFv35-MAP is rapidly internalized by target cells after binding its receptor, and exhibits specific cytotoxicity toward FL-OH1 and RD cells in vitro. We also confirmed that scFv35-MAP induces apoptosis in FL-OH1 and RD cells. The in vivo potential of scFv35-MAP will need to be considered in further studies.


Assuntos
Apoptose/efeitos dos fármacos , Antagonistas Colinérgicos/farmacologia , Proteínas Associadas aos Microtúbulos/genética , Proteínas Recombinantes de Fusão/farmacologia , Rabdomiossarcoma/tratamento farmacológico , Camptotecina/farmacologia , Linhagem Celular Tumoral , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Imunoterapia , Ligação Proteica , Receptores Colinérgicos/metabolismo , Proteínas Recombinantes de Fusão/genética , Células U937
4.
J Cancer Res Clin Oncol ; 141(6): 1049-61, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25433506

RESUMO

PURPOSE: Rhabdomyosarcoma (RMS) is a rare and aggressive soft tissue sarcoma with limited treatment options and a high failure rate during standard therapy. New therapeutic strategies based on targeted immunotherapy are therefore much in demand. The epidermal growth factor receptor (EGFR) has all the characteristics of an ideal target. It is overexpressed in up to 80 % of embryonal RMS and up to 50 % of alveolar RMS tumors. We therefore tested the activity of the EGFR-specific recombinant immunotoxin (IT) 425(scFv)-ETA' against EGFR(+) RMS cells in vitro and ex vivo. METHODS: We tested the specific binding and internalization behavior of 425(scFv)-ETA' in RMS cell lines in vitro by flow cytometry, compared to the corresponding imaging probe 425(scFv)-SNAP monitored by live cell imaging. The cytotoxic activity of 425(scFv)-ETA' was tested using cell viability and apoptosis assays. Specific binding of the IT was confirmed on formalin-fixed paraffin-embedded tissue samples from two RMS patients. RESULTS: We confirmed the specific binding of 425(scFv)-ETA' to RMS cells in vitro and ex vivo. Both the IT and the corresponding imaging probe were rapidly internalized. The IT killed EGFR(+) RMS cells in a dose-dependent manner, while showing no effect against control cells. It showed specific apoptotic activity against one selected RMS cell line. CONCLUSIONS: This is the first study showing the promising therapeutic potential of a recombinant, EGFR-targeting, ETA'-based IT on RMS cells. We confirmed the selective killing with IC50 values of up to 50 pM, and immunohistochemical staining confirmed the specific ex vivo binding to primary RMS material.


Assuntos
Receptores ErbB/metabolismo , Imunoterapia/métodos , Imunotoxinas/farmacologia , Rabdomiossarcoma/tratamento farmacológico , Rabdomiossarcoma/patologia , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Microscopia Confocal , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Rabdomiossarcoma/imunologia , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma Alveolar/tratamento farmacológico , Rabdomiossarcoma Alveolar/patologia , Rabdomiossarcoma Embrionário/tratamento farmacológico , Rabdomiossarcoma Embrionário/patologia , Resultado do Tratamento , Regulação para Cima
5.
BioDrugs ; 28(2): 181-209, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24043362

RESUMO

Hodgkin's lymphoma (HL) and ALK(+) anaplastic large-cell lymphoma (ALCL) have become highly curable due to the success of modern regimens of chemotherapy and radiotherapy. However, up to one-third of the patients experience relapse or do not respond to first-line therapy, and half of them relapse again after secondary therapy with limited options for further treatment. In the last 15 years, monoclonal antibodies (mAbs) directed to surface receptors became a new and valuable therapeutic option in many hematologic malignancies. Due to its restricted expression on normal activated lymphocytes and its high expression on malignant cells, CD30 represents an attractive target molecule for HL and ALCL therapy. However, unconjugated CD30 mAbs have demonstrated a lack of objective clinical responses in patients with recurrent HL. CD30 exhibits complex signaling pathways, and binding of its natural ligand or anti-CD30 mAbs can induce apoptosis but may also promote proliferation and activation depending on the cellular context. Moreover, CD30 rapidly internalizes after crosslinking, which counteracts efficient recruitment of immunologic effectors but also provides the opportunity to transfer cytotoxic payloads coupled to CD30-specific mAbs into the tumor cells. Several tumor targeting approaches have been studied, including radio-immunoconjugates, immunotoxins, immunoRNases, immunokinases, and antibody drug conjugates (ADCs). In 2011, the ADC brentuximab-vedotin, consisting of the CD30-specific chimeric mAb cAC10 and the potent tubulin toxin monomethyl auristatin E, gained regulatory approval as a well tolerated and highly active drug in patients with refractory and relapsed HL and ALCL. SGN-35 is on the way to being incorporated in the standard management of CD30(+) lymphoma with significant therapeutic impact. This review gives a critical overview about anti-CD30 therapies with unconjugated, engineered, and conjugated mAbs and the therapeutic challenges of treatment of CD30(+) lymphoma.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Doença de Hodgkin/tratamento farmacológico , Antígeno Ki-1/antagonistas & inibidores , Linfoma Anaplásico de Células Grandes/tratamento farmacológico , Quinase do Linfoma Anaplásico , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Doença de Hodgkin/imunologia , Doença de Hodgkin/patologia , Humanos , Antígeno Ki-1/imunologia , Linfoma Anaplásico de Células Grandes/imunologia , Linfoma Anaplásico de Células Grandes/patologia , Terapia de Alvo Molecular , Receptores Proteína Tirosina Quinases/genética
6.
Artigo em Inglês | MEDLINE | ID: mdl-19149683

RESUMO

RNA interference (RNAi) is a powerful endogenous process initiated by short double stranded RNAs, which results in sequence-specific posttranscriptional gene silencing. The ability to block the expression of any disease-causing or disease-related protein emphasizes the huge therapeutic potential of this technology. In a clinical setting, however, the use of RNAi-based therapeutics is limited by their short serum half lives and poor uptake into cells. In this review, we provide an overview of recent patents in the field of short interfering RNA (siRNA) delivery and discuss recent progress in the development of efficient siRNA delivery vehicles enhancing the pharmacokinetic properties of RNAi-based therapeutics and promoting cellular uptake.


Assuntos
Sistemas de Liberação de Medicamentos , RNA Interferente Pequeno/administração & dosagem , Animais , Aptâmeros de Nucleotídeos/administração & dosagem , Humanos , Interferência de RNA , RNA Interferente Pequeno/química , RNA Interferente Pequeno/farmacocinética , Distribuição Tecidual
7.
Mol Cancer Ther ; 7(9): 2924-32, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18790773

RESUMO

Acute myeloid leukemia (AML) cells of subtypes M4 and M5 show enhanced expression of CD64 (FcgammaRI), the high-affinity receptor for IgG, which is normally expressed at high levels only on activated cells of the myeloid lineage. CD64 is therefore a prime target for the specific delivery of cytotoxic agents. A promising toxin candidate is granzyme B, a human serine protease originating from cytotoxic granules of CD8+ T lymphocytes and natural killer cells. After evaluating the sensitivity of the AML-related cell line U937 toward cytosolic granzyme B, we genetically fused granzyme B to H22, a humanized single-chain antibody fragment (scFv) specific for CD64, to obtain Gb-H22(scFv), a fusion protein lacking the immunogenic properties of nonhuman immunofusions. Gb-H22(scFv) was successfully expressed in human 293T cells, secreted, and purified from cell culture supernatants. The purified protein bound specifically to CD64+ U937 cells. Despite linkage to the binding domain, the proteolytic activity of functional Gb-H22(scFv) was identical to that of free granzyme B. Target cell-specific cytotoxicity was observed with a half-maximal inhibitory concentration (IC50) between 1.7 and 17 nmol/L. In addition, the induction of apoptosis in U937 cells was confirmed by Annexin A5 staining and the detection of activated caspase-3 in the cytosol. Finally, apoptosis was observed in primary CD64+ AML cells, whereas CD64(-) AML cells were unaffected. This is the first report of a completely human granzyme B-based immunotoxin directed against CD64, with activity against an AML-related cell line and primary AML cells.


Assuntos
Granzimas/farmacologia , Região Variável de Imunoglobulina/farmacologia , Imunotoxinas/farmacologia , Leucemia Monocítica Aguda/metabolismo , Receptores de IgG/antagonistas & inibidores , Proteínas Recombinantes de Fusão/farmacologia , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Ativação Enzimática/efeitos dos fármacos , Humanos , Leucemia Monocítica Aguda/enzimologia , Leucemia Monocítica Aguda/imunologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Células U937
8.
Biochem Biophys Res Commun ; 338(4): 1811-7, 2005 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-16289041

RESUMO

The ectodomain of human FcgammaRI (rsCD64) was expressed in HEK 293T cells and purified by immobilized-metal affinity chromatography. Binding activity to human IgG was verified by ELISA and the isotype-specificity determined by a surface plasmon resonance inhibition assay was found to be the same as for native CD64. The active concentration of the rsCD64 preparation was derived using a solution competition assay and was used for the subsequent kinetic analysis. Binding curves were well described by a simple monovalent interaction model confirming the known stoichiometry of the interaction. Mass-transport limitation was prevented by using sufficiently low surface capacities. For binding to the recombinant mouse/human chimeric antibody cPIPP (IgG1/kappa) a high association rate of k(ass)=1.7 x 10(6) (M s)(-1) and a low dissociation rate of k(diss)=1.8 x 10(-4) s(-1) were observed. The derived dissociation equilibrium constant of K(D)=110 pM was significantly lower than that reported for binding to native FcgammaRI.


Assuntos
Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Receptores de IgG/imunologia , Receptores de IgG/metabolismo , Animais , Reações Antígeno-Anticorpo , Linhagem Celular , Quimera/imunologia , Cromatografia de Afinidade , Ensaio de Imunoadsorção Enzimática , Humanos , Cinética , Camundongos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Ressonância de Plasmônio de Superfície
9.
Int J Mol Med ; 14(4): 765-8, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15375615

RESUMO

Due to problems of immobilizing functional tumor antigens in their natural conformation on surfaces for immunoassays, it is often difficult to evaluate the binding of antibodies derived from phage display libraries depleted and selected by panning on cell lines and living tumor cells. Performing cell membrane based ELISA methods does not reveal any up front kinetic binding information and depends on the performance of secondary antibodies and substrates. To overcome these limitations, we developed a new method to visualize direct antibody-cell membrane interactions by surface plasmon resonance using the Biacore 3000 and on-line signal subtraction on antigen-negative cell membrane vesicles. Conditions for the coating of cell membrane preparations to a carboxymethyl dextran hydrogel surface of a commercially available chip and the proof of concept for this application by the analysis of different formats of anti-CD30 and anti-carcinoembryonic antigen (CEA) antibodies interacting with coated membrane vesicles of CD30-positive/CEA-negative and CD30-negative and CEA-positive cell lines are described.


Assuntos
Anticorpos/imunologia , Anticorpos/metabolismo , Membrana Celular/imunologia , Membrana Celular/metabolismo , Ressonância de Plasmônio de Superfície/métodos , Linhagem Celular Tumoral , Humanos , Antígeno Ki-1/imunologia , Antígeno Ki-1/metabolismo
10.
Int J Cancer ; 111(4): 568-74, 2004 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-15239135

RESUMO

There is increasing evidence that the shedding of extracellular antigen domains impedes selective immunotherapy. One example is CD30, which is overexpressed on the surface of malignant lymphoma cells and has been identified as a promising target for antibody-based immunotherapy. However, CD30 is cleaved from the surface of target cells and the resulting soluble ectodomain (sCD30) is protecting the cells from antibody binding. Shedding can be inhibited by hydroxamate inhibitors of metalloproteinases such as BB-3644. We thus evaluated the influence of BB-3644 on the efficacy of the anti-CD30 single-chain immunotoxin Ki-3(scFv)-ETA'. In vitro, the addition of BB-3644 augmented the antitumor effect of Ki-3(scFv)-ETA' against Hodgkin-derived L540Cy cells by a factor of 2.75. Severe combined immunodeficiency (SCID) mice challenged with CD30-positive L540Cy cells were treated with the immunotoxin. One single nontoxic dose of BB-3644 increased the mean survival time of animals treated concomitantly with Ki-3(scFv)-ETA' to 93 days as compared with 35 days in the control (p = 0.0017). When BB-3644 was continuously delivered using subcutaneously implanted pumps, this effect was even more pronounced with no observed tumor growth in the animals within 200 days. Thus, concomitant application of metalloproteinase inhibitors might become clinically relevant in antibody-based immunotherapy against targets known to be shed from tumor cells.


Assuntos
Aminopiridinas/farmacologia , Doença de Hodgkin/patologia , Ácidos Hidroxâmicos/farmacologia , Imunoterapia/métodos , Imunotoxinas/farmacologia , Antígeno Ki-1/imunologia , Metaloendopeptidases/antagonistas & inibidores , Animais , Anticorpos , Camundongos , Camundongos SCID , Anticorpos de Cadeia Única , Análise de Sobrevida , Células Tumorais Cultivadas
11.
Cancer Res ; 63(23): 8414-9, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14679004

RESUMO

CD64, the high affinity receptor for IgG (FcgammaRI) is expressed on acute myeloid leukemia blast cells and has recently been described as a specific target for immunotherapy. To generate a recombinant immunotoxin, the anti-CD64 single chain fragment (scFv) m22 was cloned into the bacterial expression vector pBM1.1 and fused to a deletion mutant of Pseudomonas exotoxin A (ETA'). Genetically modified Escherichia coli BL21 Star (DE3) were grown under osmotic stress conditions in the presence of compatible solutes. After isopropyl beta-D-thiogalactoside induction, the 70-kDa His(10)-tagged m22(scFv)-ETA' was directed into the periplasmic space and purified by a combination of metal-ion affinity and molecular size-chromatography. The characteristics of the recombinant protein were assessed by ELISA, flow cytometry, and toxicity assays, using CD64-positive AML cells. Binding specificity of m22(scFv)-ETA' was verified by competition with the parental anti-CD64 monoclonal antibody m22. The recombinant immunotoxin showed significant toxicity toward the CD64-positive cell lines HL-60 and U937 reaching 50% inhibition of cell proliferation at a concentration (IC(50)) of 11.6 ng/ml against HL-60 cells and 12.9 ng/ml against U937 cells. Approximately 41% of primary leukemia cells from a patient with CD64-positive AML were driven into early apoptosis by m22(scFv)-ETA' as measured by flow cytometric analysis. This is the first article documenting the specific cytotoxicity of a novel recombinant immunotoxin with major implications for immunotherapy of CD64-positive diseases.


Assuntos
ADP Ribose Transferases/farmacologia , Toxinas Bacterianas/farmacologia , Exotoxinas/farmacologia , Imunotoxinas/farmacologia , Leucemia Mieloide/tratamento farmacológico , Receptores de IgG/imunologia , Fatores de Virulência/farmacologia , ADP Ribose Transferases/genética , ADP Ribose Transferases/imunologia , Adulto , Especificidade de Anticorpos , Apoptose/efeitos dos fármacos , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Exotoxinas/genética , Exotoxinas/imunologia , Humanos , Fragmentos de Imunoglobulinas/genética , Fragmentos de Imunoglobulinas/imunologia , Fragmentos de Imunoglobulinas/farmacologia , Imunotoxinas/genética , Imunotoxinas/imunologia , Leucemia Mieloide/imunologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Exotoxina A de Pseudomonas aeruginosa
12.
Protein Expr Purif ; 28(2): 211-9, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12699683

RESUMO

Immunotoxins consist of a target-cell-specific binding moiety, chemically or recombinantly linked to a cytotoxic component. A number of different immunotoxins (IT) have increasingly been evaluated for immunotherapy. Since these foreign proteins are highly immunogenic in human, we have developed recombinant IT using the human ribonuclease angiogenin. Due to their potential toxic effects on eucaryotic cells, these IT are usually expressed in bacteria. Depending on the structure, size, and sequence of the desired IT, bacterial expression can be limited and the yield after purification be unsatisfactory. Therefore, the expression of IT in eucaryotic cells could provide a promising alternative. For this purpose we genetically fused the anti-CD30 single-chain variable fragment (scFv) Ki4 to the N- and C-termini of recombinant angiogenin. Both IT possess leader sequences, which mediate their secretion into the cell culture supernatant. Using a bicistronic mRNA the IT were simultaneously expressed together with enhanced green fluorescent protein (EGFP). This allows direct monitoring of transfected cells. An additional plasmid encoded Zeocin resistance enhances the cultivation of transfected cells under selection pressure. Three days after transfection of 293T-cells, unpurified IT were analyzed by flow cytometry and competitive cell proliferation assays. This is the first report on the use of eucaryotic cells for the secretion of functionally active IT with a human effector domain.


Assuntos
Anticorpos Monoclonais/genética , Imunotoxinas/metabolismo , Antígeno Ki-1/imunologia , Ribonuclease Pancreático/genética , Anticorpos Monoclonais/imunologia , Western Blotting , Linhagem Celular , Meios de Cultivo Condicionados/metabolismo , Eletroforese em Gel de Poliacrilamida , Células Eucarióticas/metabolismo , Citometria de Fluxo , Humanos , Fragmentos de Imunoglobulinas/genética , Fragmentos de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Imunotoxinas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...