Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Oncoimmunology ; 7(9): e1472195, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30228941

RESUMO

A number of agents designed for immunotherapy of Acute Myeloid Leukemia (AML) are in preclinical and early clinical development. Most of them target a single antigen on the surface of AML cells. Here we describe the development and key biological properties of a tri-specific agent, the dual-targeting triplebody SPM-2, with binding sites for target antigens CD33 and CD123, and for CD16 to engage NK cells as cytolytic effectors. Primary blasts of nearly all AML patients carry at least one of these target antigens and the pair is particularly promising for the elimination of blasts and leukemia stem cells (LSCs) from a majority of AML patients by dual-targeting agents. The cytolytic activity of NK cells mediated by SPM-2 was analyzed in vitro for primary leukemic cells from 29 patients with a broad range of AML-subtypes. Blasts from all 29 patients, including patients with genomic alterations associated with an unfavorable genetic subtype, were lysed at nanomolar concentrations of SPM-2. Maximum susceptibility was observed for cells with a combined density of CD33 and CD123 above 10,000 copies/cell. Cell populations enriched for AML-LSCs (CD34pos and CD34pos CD38neg cells) from 2 AML patients carried an increased combined antigen density and were lysed at correspondingly lower concentrations of SPM-2 than unsorted blasts. These initial findings raise the expectation that SPM-2 may also be capable of eliminating AML-LSCs and thus of prolonging survival. In the future, patients with a broad range of AML subtypes may benefit from treatment with SPM-2.

2.
Oncotarget ; 7(50): 83392-83408, 2016 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-27825135

RESUMO

Triplebodies are antibody-derived recombinant proteins carrying 3 antigen-binding domains in a single polypeptide chain. Triplebody SPM-1 was designed for lysis of CD19-bearing malignant B-lymphoid cells through the engagement of CD16-expressing cytolytic effectors, including NK and γδ T cells.SPM-1 is an optimized version of triplebody ds(19-16-19) and includes humanization, disulfide stabilization and the removal of potentially immunogenic sequences. A three-step chromatographic procedure yielded 1.7 - 5.5 mg of purified, monomeric protein per liter of culture medium. In cytolysis assays with NK cell effectors, SPM-1 mediated potent lysis of cancer-derived B cell lines and primary cells from patients with various B-lymphoid malignancies, which surpassed the ADCC activity of the therapeutic antibody Rituximab. EC50-values ranged from 3 to 86 pM. Finally, in an impedance-based assay, SPM-1 mediated a particularly rapid lysis of CD19-bearing target cells by engaging and activating both primary and expanded human γδ T cells from healthy donors as effectors.These data establish SPM-1 as a useful tool for a kinetic analysis of the cytolytic reactions mediated by γδ T and NK cells and as an agent deserving further development towards clinical use for the treatment of B-lymphoid malignancies.


Assuntos
Antígenos CD19/imunologia , Antineoplásicos Imunológicos/farmacologia , Citotoxicidade Imunológica/efeitos dos fármacos , Linfócitos Intraepiteliais/efeitos dos fármacos , Células Matadoras Naturais/efeitos dos fármacos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfoma de Células B/tratamento farmacológico , Antineoplásicos Imunológicos/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Linfócitos Intraepiteliais/imunologia , Células Matadoras Naturais/imunologia , Cinética , Ativação Linfocitária/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Linfoma de Células B/imunologia , Linfoma de Células B/patologia , Rituximab/farmacologia , Células Tumorais Cultivadas
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.
Analyst ; 141(7): 2284-95, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-26958659

RESUMO

Cancer therapy via redirected lysis mediated by antibodies and antibody-derived agents relies on the availability of substantial numbers of sufficiently active immune effector cells. To monitor antitumor responses before and during therapy, sensitive methods are needed, capable of quantitating specific lysis of target cells. Here we present a chip-based single-cell cytometric assay, which uses adherent human target cells arrayed in structured micro-fields. Using a fluorescent indicator of cell death and time-lapse microscopy in an automated high-throughput mode, we measured specific target cell lysis by activated human NK cells, mediated by the therapeutic single chain triplebody SPM-2 (33-16-123). This antibody-derived tri-specific fusion protein carries binding sites for the myeloid antigens CD33 and CD123 and recruits NK cells via a binding site for the Fc-receptor CD16. Specific lysis increased with increasing triplebody concentration, and the single-cell assay was validated by direct comparison with a standard calcein-release assay. The chip-based approach allowed measurement of lysis events over 16 hours (compared to 4 hours for the calcein assay) and required far smaller numbers of primary cells. In addition, dynamic properties inaccessible to conventional methods provide new details about the activation of cytolytic effector cells by antibody-derived agents. Thus, the killing rate exhibited a dose-dependent maximum during the reaction interval. In clinical applications ex vivo monitoring of NK activity of patient's endogenous cells will likely help to choose appropriate therapy, to detect impaired or recovered NK function, and possibly to identify rare subsets of cancer cells with particular sensitivity to effector-cell mediated lysis.


Assuntos
Células Matadoras Naturais/citologia , Procedimentos Analíticos em Microchip/métodos , Análise de Célula Única/métodos , Anticorpos de Cadeia Única/metabolismo , Morte Celular , Linhagem Celular Tumoral , Células HEK293 , Humanos , Fatores de Tempo
5.
Oncotarget ; 7(16): 22579-89, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-26981773

RESUMO

Simultaneous targeting of multiple tumor-associated antigens (TAAs) in cancer immunotherapy is presumed to enhance tumor cell selectivity and to reduce immune escape.The combination of B lymphoid marker CD19 and myeloid marker CD33 is exclusively present on biphenotypic B/myeloid leukemia cells. Triplebody 33-3-19 binds specifically to both of these TAAs and activates T cells as immune effectors. Thereby it induces specific lysis of established myeloid (MOLM13, THP-1) and B-lymphoid cell lines (BV173, SEM, Raji, ARH77) as well as of primary patient cells. EC50 values range from 3 pM to 2.4 nM. In accordance with our hypothesis, 33-3-19 is able to induce preferential lysis of double- rather than single-positive leukemia cells in a target cell mixture: CD19/CD33 double-positive BV173 cells were eliminated to a significantly greater extent than CD19 single-positive SEM cells (36.6% vs. 20.9% in 3 hours, p = 0.0048) in the presence of both cell lines. In contrast, equivalent elimination efficiencies were observed for both cell lines, when control triplebody 19-3-19 or a mixture of the bispecific single chain variable fragments 19-3 and 33-3 were used. This result highlights the potential of dual-targeting agents for efficient and selective immune-intervention in leukemia patients.


Assuntos
Antígenos de Neoplasias/efeitos dos fármacos , Antineoplásicos/farmacologia , Imunoterapia/métodos , Leucemia Aguda Bifenotípica , Anticorpos de Cadeia Única/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígenos CD19/imunologia , Antígenos de Diferenciação Mielomonocítica/efeitos dos fármacos , Antígenos de Diferenciação Mielomonocítica/imunologia , Humanos , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia
6.
Cancer Immunol Immunother ; 64(12): 1623-35, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26515978

RESUMO

Chimeric antigen receptor (CAR)-modified T cells emerged as effective tools in the immunotherapy of cancer but can produce severe on-target off-tissue toxicities. This risk can conceivably be overcome, at least partially, by transient transfection. The design of CARs, however, has so far not been optimized for use in non-permanent T cell modification. Here we compared the performance of T cells modified with three different first- and second-generation CARs, each specific for MCSP (HMW-MAA) which is commonly expressed by melanoma cells. Upon RNA transfer, the expression of all receptors was limited in time. The second-generation CARs, which combined CD28-CD3ζ signaling, were expressed at higher levels and more prolonged than first-generation CARs with CD3ζ only. The CD28 domain increased the cytokine production, but had only an indirect effect on the lytic capacity, by prolonging the CAR expression. Especially for the second-generation CARs, the scFv clearly impacted the level and duration of CAR expression and the T cell performance. Thus, we identified a CAR high in both expression and anti-tumor cell reactivity. T cells transfected with this CAR increased the mean survival time of mice after challenge with melanoma cells. To facilitate clinical application, this CAR was used to redirect T cells from late-stage melanoma patients by RNA transfection. These T cells mediated effective antigen-specific tumor cell lysis and release of pro-inflammatory cytokines, even after cryoconservation of the transfected T cells. Taken together, the analysis identified a CAR with superior anti-melanoma performance after RNA transfer which is a promising candidate for clinical exploration.


Assuntos
Proteínas Mitocondriais/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Proteínas de Transporte/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Melanoma/imunologia , Melanoma/fisiopatologia , Camundongos , Proteínas Mitocondriais/genética , Estabilidade Proteica , Estrutura Terciária de Proteína , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Anticorpos de Cadeia Única/metabolismo
7.
Haematologica ; 100(3): 336-44, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25480499

RESUMO

Current therapies for acute myeloid leukemia are associated with high failure and relapse rates. Adoptive immunotherapies, which have shown promise in the treatment of hematologic malignancies, have the potential to target acute myeloid leukemia through pathways that are distinct and complementary to current approaches. Here, we describe the development of a novel adoptive immunotherapy specific for this disease. We generated a second generation CD33-specific chimeric antigen receptor capable of redirecting cytolytic effector T cells against leukemic cells. CD33 is expressed in approximately 90% of acute myeloid leukemia cases and has demonstrated utility as a target of therapeutic antibodies. Chimeric antigen receptor-modified T cells efficiently killed leukemia cell lines and primary tumor cells in vitro. The anti-leukemia effect was CD33-specific, mediated through T-cell effector functions, and displayed tumor lysis at effector:target ratios as low as 1:20. Furthermore, the CD33-redirected T cells were effective in vivo, preventing the development of leukemia after prophylactic administration and delaying the progression of established disease in mice. These data provide pre-clinical validation of the effectiveness of a second-generation anti-CD33 chimeric antigen receptor therapy for acute myeloid leukemia, and support its continued development as a clinical therapeutic.


Assuntos
Imunoterapia Adotiva/métodos , Leucemia Mieloide Aguda/terapia , Proteínas Mutantes Quiméricas/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/antagonistas & inibidores , Linfócitos T Citotóxicos/imunologia , Animais , Medula Óssea/imunologia , Medula Óssea/patologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Citotoxicidade Imunológica , Expressão Gênica , Humanos , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/imunologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/patologia , Fígado/imunologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Mutantes Quiméricas/genética , Plasmídeos/química , Plasmídeos/genética , Receptores de Antígenos de Linfócitos T/genética , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Baço/imunologia , Baço/patologia , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/transplante , Transfecção
8.
Oncotarget ; 5(15): 6466-83, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25115385

RESUMO

Triplebody 19-3-19, an antibody-derived protein, carries three single chain fragment variable domains in tandem in a single polypeptide chain. 19-3-19 binds CD19-bearing lymphoid cells via its two distal domains and primary T cells via its CD3-targeting central domain in an antigen-specific manner. Here, malignant B-lymphoid cell lines and primary cells from patients with B cell malignancies were used as targets in cytotoxicity tests with pre-stimulated allogeneic T cells as effectors. 19-3-19 mediated up to 95 % specific lysis of CD19-positive tumor cells and, at picomolar EC50 doses, had similar cytolytic potency as the clinically successful agent Blinatumomab. 19-3-19 activated resting T cells from healthy unrelated donors and mediated specific lysis of both autologous and allogeneic CD19-positive cells. 19-3-19 led to the elimination of 70 % of CD19-positive target cells even with resting T cells as effectors at an effector-to-target cell ratio of 1 : 10. The molecule is therefore capable of mediating serial lysis of target cells by a single T cell. These results highlight that central domains capable of engaging different immune effectors can be incorporated into the triplebody format to provide more individualized therapy tailored to a patient's specific immune status.


Assuntos
Linfócitos/imunologia , Linfoma de Células B/terapia , Anticorpos de Cadeia Única/farmacologia , Linfócitos T/imunologia , Idoso , Idoso de 80 Anos ou mais , Citotoxicidade Celular Dependente de Anticorpos , Feminino , Células HEK293 , Humanos , Imunização Passiva/métodos , Ativação Linfocitária , Linfoma de Células B/imunologia , Masculino , Pessoa de Meia-Idade , Anticorpos de Cadeia Única/imunologia , Adulto Jovem
9.
Cancer Lett ; 352(2): 228-35, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25016058

RESUMO

The treatment of rhabdomyosarcoma (RMS) remains challenging, with metastatic and alveolar RMS offering a particularly poor prognosis. Therefore, the identification and evaluation of novel antigens, which are suitable targets for immunotherapy, is one attractive possibility to improve the treatment of this disease. Here we show that chondroitin sulfate proteoglycan 4 (CSPG4) is expressed on RMS cell lines and RMS patient material. We evaluated the immunotoxin (IT) αMCSP-ETA', which specifically recognizes CSPG4 on the RMS cell lines RD, FL-OH1, TE-671 and Rh30. It is internalized rapidly, induces apoptosis and thus kills RMS cells selectively. We also demonstrate the specific binding of this IT to RMS primary tumor material from three different patients.


Assuntos
ADP Ribose Transferases/farmacologia , Apoptose/efeitos dos fármacos , Toxinas Bacterianas/farmacologia , Proteoglicanas de Sulfatos de Condroitina/imunologia , Exotoxinas/farmacologia , Imunotoxinas/farmacologia , Proteínas de Membrana/imunologia , Rabdomiossarcoma/patologia , Anticorpos de Cadeia Única/farmacologia , Fatores de Virulência/farmacologia , ADP Ribose Transferases/metabolismo , Toxinas Bacterianas/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Relação Dose-Resposta a Droga , Exotoxinas/metabolismo , Humanos , Imunotoxinas/imunologia , Imunotoxinas/metabolismo , Concentração Inibidora 50 , Proteínas de Membrana/metabolismo , Ligação Proteica , Rabdomiossarcoma/imunologia , Rabdomiossarcoma/metabolismo , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/metabolismo , Fatores de Tempo , Fatores de Virulência/metabolismo , Exotoxina A de Pseudomonas aeruginosa
10.
MAbs ; 6(1): 286-96, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24135631

RESUMO

The single-chain triplebody HLA-ds16-hu19 consists of three single-chain Fv (scFv) antibody fragments connected in a single polypeptide chain. This protein with dual-targeting capacity mediated preferential lysis of antigen double positive(dp) over single-positive (sp) leukemic cells by recruitment of natural killer (NK) cells as effectors. The two distal scFv modules were specific for the histocompatibility protein HLA-DR and the lymphoid antigen CD19, the central one for the Fc gamma receptor CD16. In antibody-dependent cellular cytotoxicity (ADCC) experiments with a mixture of leukemic target cells comprising both HLA-DR sp HuT-78 or Kasumi-1 cells and (HLA-DR plus CD19) dp SEM cells, the triplebody mediated preferential lysis of the dp cells even when the sp cells were present in ≤ 20-fold numerical excess.The triplebody promoted equal lysis of SEM cells at 2.5-fold and 19.5-fold lower concentrations than the parental antibodies specific for HLA-DR and CD19, respectively. Finally, the triplebody also eliminated primary leukemic cells at lower concentrations than an equimolar mixture of bispecific single-chain Fv fragments (bsscFvs) separately addressing each target antigen (hu19-ds16 and HLA-ds16). The increased selectivity of targeting and the preferential lysis of dp over sp cells achieved by dual-targeting open attractive new perspectives for the use of dual-targeting agents in cancer therapy.


Assuntos
Anticorpos Antineoplásicos/farmacologia , Antígenos CD19/imunologia , Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos , Antígenos HLA-DR/imunologia , Imunidade Celular/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Proteínas de Neoplasias/imunologia , Anticorpos de Cadeia Única/farmacologia , Animais , Anticorpos Antineoplásicos/genética , Anticorpos Antineoplásicos/imunologia , Antígenos CD19/genética , Antineoplásicos/imunologia , Células CHO , Cricetinae , Cricetulus , Feminino , Antígenos HLA-DR/genética , Humanos , Imunidade Celular/imunologia , Células Matadoras Naturais/patologia , Masculino , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia
11.
J Transl Med ; 11: 289, 2013 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-24237598

RESUMO

BACKGROUND: The capacity of patient's Natural Killer cells (NKs) to be activated for cytolysis is an important prerequisite for the success of antibody-derived agents such as single-chain triplebodies (triplebodies) in cancer therapy. NKs recovered from AML patients at diagnosis are often found to be reduced in peripheral blood titers and cytolytic activity. Here, we had the unique opportunity to compare blood titers and cytolytic function of NKs from an AML patient with those of a healthy monozygotic twin. The sibling's NKs were compared with the patient's drawn either at diagnosis or in remission after chemotherapy. The cytolytic activities of NKs from these different sources for the patient's autologous AML blasts and other leukemic target cells in conjunction with triplebody SPM-2, targeting the surface antigens CD33 and CD123 on the AML cells, were compared. METHODS: Patient NKs drawn at diagnosis were compared to NKs drawn in remission after chemotherapy and a sibling's NKs, all prepared from PBMCs by immunomagnetic beads (MACS). Redirected lysis (RDL) assays using SPM-2 and antibody-dependent cellular cytotoxicity (ADCC) assays using the therapeutic antibody RituximabTM were performed with the enriched NKs. In addition, MACS-sorted NKs were analyzed for NK cell activating receptors (NCRs) by flow cytometry, and the release of TNF-alpha and IFN-gamma from blood samples of both siblings after the addition of the triplebody were measured in ELISA-assays. RESULTS: Patient NKs isolated from peripheral blood drawn in remission produced comparable lysis as NKs from the healthy twin against the patient's autologous bone marrow (BM) blasts, mediated by SPM-2. The NCR receptor expression profiles on NKs from patient and twin were similar, but NK cell titers in peripheral blood were lower for samples drawn at diagnosis than in remission. CONCLUSIONS: Peripheral blood NK titers and ex vivo cytolytic activities mediated by triplebody SPM-2 were comparable for cells drawn from an AML patient in remission and a healthy twin. If these results can be generalized, then NKs from AML patients in remission are sufficient in numbers and cytolytic activity to make triplebodies promising new agents for the treatment of AML.


Assuntos
Citotoxicidade Imunológica , Células Matadoras Naturais/imunologia , Leucemia Mieloide Aguda/imunologia , Indução de Remissão , Gêmeos Monozigóticos , Adulto , Citotoxicidade Celular Dependente de Anticorpos , Feminino , Citometria de Fluxo , Humanos , Adulto Jovem
12.
MAbs ; 4(1): 45-56, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22327429

RESUMO

To test the hypothesis that dual-targeting confers the novel ability of selective binding to antigen double-positive over antigen single-positive cells, a single-chain triplebody (sctb), HLA-ds16-hu19, was produced and characterized. The molecule carries three single-chain Fv (scFv) antibody fragments in a single polypeptide chain, the two distal ones specific for the human histocompatibility protein HLA-DR and the B-lymphoid cell surface protein CD19, the central one for CD16, the human low affinity Fc-receptor FcγRIII. For comparison, the bispecific scFvs (bsscFv) hu19-ds16 and HLA-ds16 were also produced. All CD16 binding modules are disulfide-stabilized (ds). The sctb bound simultaneously to both CD19 and HLA-DR on the same cancer cell and, thus, showed functional dual-targeting. In a mixing-experiment with HLA-DR single-positive HUT-78 cells and (HLA-DR plus CD19) double-positive SEM cells, the triplebody showed preferential binding to the double-positive cells, even when the single-positive cells were present in a numerical excess of up to 20-fold. In antibody-dependent cellular cytotoxicity experiments with mononuclear cells as effector cells, the sctb promoted equal lysis of Raji cells, an antigen double-positive cell line, at 130-fold lower concentrations than the bsscFv hu19-ds16, indicating that both distal scFvs of the sctb contributed to tumor cell lysis. A panel of stably-transfected HEK293 cell lines was generated that included CD19- and HLA-DR single-positive and (HLA-DR plus CD19) double-positive lines with antigen-surface densities varying over a broad range. Using a pair of cell lines with matching densities, the sctb eliminated double-positive target cells preferentially single-positive cells. This ability of preferential or selective targeting of antigen double-positive over single-positive cells opens attractive new perspectives for the use of dual-targeting sctbs in cancer therapy.


Assuntos
Anticorpos Biespecíficos/imunologia , Especificidade de Anticorpos/imunologia , Antígenos CD19/imunologia , Antígenos HLA-DR/imunologia , Receptores de IgG/imunologia , Proteínas Recombinantes de Fusão/imunologia , Animais , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/metabolismo , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígenos CD19/metabolismo , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Proteínas Ligadas por GPI/imunologia , Proteínas Ligadas por GPI/metabolismo , Células HEK293 , Antígenos HLA-DR/metabolismo , Humanos , Células Matadoras Naturais/imunologia , Ligação Proteica , Receptores de IgG/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/metabolismo
13.
J Immunol Methods ; 373(1-2): 67-78, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-21855548

RESUMO

Protein- or glyco-engineering of antibody molecules can be used to enhance Fc-mediated effector functions. ScFv-Fc fusion proteins (scFv-Fc) represent interesting antibody derivatives due to their relatively simple design and increased tissue penetration. Here, the impact of protein- and glyco-engineering on ADCC potency of a panel of human IgG1-based scFv-Fc was tested. Three matched sets of scFv-Fc variants targeting CD7, CD20 or HLA class II and optimized for CD16a binding by mutagenesis, lack of core-fucose, or their combination, were generated and functionally tested in comparison to the corresponding wild type scFv-Fc. Antigen binding activity was not compromised by altered glycosylation or Fc mutagenesis, whereas Fc binding to CD16a was significantly enhanced in the order: non-core fucosylated/Fc-mutated double-engineered≫Fc-mutated≥non-core-fucosylated>wild-type IgG1-Fc. All engineered variants triggered potent ADCC with up to 100-fold reduced EC50 values compared to non-engineered variants. Interestingly, double-engineered variants were similarly effective in triggering ADCC compared to single-engineered variants irrespective of their 1 log greater CD16a binding affinity. Thus, these data demonstrate that protein- and glyco-engineering enhances NK-cell mediated ADCC of scFv-Fc similarly and show that enhancing CD16a affinity beyond a certain threshold does not result in a further increase of NK-cell mediated ADCC.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos/imunologia , Fragmentos Fc das Imunoglobulinas/imunologia , Células Matadoras Naturais/imunologia , Receptores de IgG/imunologia , Anticorpos de Cadeia Única/imunologia , Animais , Afinidade de Anticorpos/imunologia , Antígenos CD20/imunologia , Antígenos CD20/metabolismo , Antígenos CD7/imunologia , Antígenos CD7/metabolismo , Western Blotting , Células CHO , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Citometria de Fluxo , Fucose/metabolismo , Glicoproteínas/genética , Glicoproteínas/imunologia , Glicoproteínas/metabolismo , Antígenos HLA-D/imunologia , Antígenos HLA-D/metabolismo , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulina G/imunologia , Células Matadoras Naturais/metabolismo , Mutagênese , Ligação Proteica , Engenharia de Proteínas/métodos , Receptores Fc/imunologia , Receptores Fc/metabolismo , Receptores de IgG/genética , Receptores de IgG/metabolismo , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/metabolismo
14.
Cancer Lett ; 303(2): 128-39, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21339041

RESUMO

Bispecific scFv antibody-derivatives (bsscFvs) recruiting natural killer (NK) cells for the lysis of malignant cells have therapeutic potential. However, a bsscFv specific for the B-lymphoid tumor antigen CD19 and the trigger molecule CD16 on NK cells had similar affinities for both antigens (42 and 58nM, respectively) and was not optimal for cytotoxicity. Therefore, a bispecific tribody (bsTb) was constructed with two binding sites for CD19 and one for CD16. This bsTb contained a CD19-specific Fab fragment carrying a CD16-specific scFv fused to its light chain and a CD19-specific scFv fused to its heavy chain. The bsTb was compared with a bispecific bibody (bsBb) lacking the CD19-specific scFv. The bsTb had 3-fold greater avidity for CD19 than the bsBb (8 and 24nM, respectively), while both had equal affinity for CD16 (56nM). Both molecules mediated antibody-dependent cellular cytotoxicity (ADCC) of leukemia-derived SEM cells and primary cells from leukemia patients. The bsTb showed half-maximum effective concentrations (EC(50)) of 55pM and promoted equal lysis as the bsBb and the bsscFv at 6- and 12-fold lower concentrations, respectively. Among these three molecules the bsTb showed the most promising in vitro properties which are anticipated to be displayed also in vivo.


Assuntos
Anticorpos Biespecíficos/química , Antígenos CD19/química , Linfoma de Células B/imunologia , Receptores de IgG/química , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Linhagem Celular Tumoral , Dimerização , Citometria de Fluxo/métodos , Humanos , Fragmentos de Imunoglobulinas , Imunoterapia/métodos , Cinética , Leucócitos Mononucleares/citologia , Linfoma de Células B/terapia , Ligação Proteica , Proteínas Recombinantes de Fusão/química
15.
MAbs ; 3(1): 21-30, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21081841

RESUMO

A single-chain triplebody (sctb) 33-ds16-ds19 comprising two distal single-chain Fv fragments (scFvs) specific for the lymphoid antigen CD19 and the myeloid antigen CD33 flanking a central scFv specific for CD16, which is the low affinity Fc-receptor (FcγRIII) present on natural killer cells and macrophages, was produced and its properties were investigated. CD33 and CD19 in combination are present on acute leukemiablasts with mixed lineage phenotype, but not on normal human hematopoietic cells. For comparison, two bispecific scFvs (bsscFvs), ds19-ds16 and 33-ds16, with monovalent binding to CD19 and CD33, respectively, were also studied. The sctb 33-ds16-ds19 specifically interacted with all 3 antigens. On the antigen double-positive cell line BV-173, the sctb bound with 2-fold greater avidity than bsscFv ds19-ds16 (KD = 21 vs. 42 nM) and with 1.4-fold greater avidity than bsscFv 33-ds16 (KD = 29 nM). All 3 fusion proteins had similar affinity for CD16 and sufficient thermic stability in human serum. In antibody-dependent cellular cytotoxicity (ADCC) reactions with human mononuclear cells as effectors, the sctb promoted lysis of BV-173 cells at 23-fold lower concentrations than bsscFv ds19-ds16 and at 1.4-fold lower concentrations than bsscFv 33-ds16. The sctb also mediated potent ADCC of the antigen double-positive mixed lineage leukemia cell line SEM, and the half-maximal concentration EC50 for BV-173 cells was 7 pM. Therefore, CD19 and CD33 are present on the surface of these leukemic cell lines such that they can be connected by a single sctb molecule, permitting the recruitment of NK cells via CD16 and tumor cell lysis.


Assuntos
Antígenos CD19/imunologia , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Anticorpos de Cadeia Única/imunologia , Animais , Especificidade de Anticorpos/imunologia , Antígenos CD/metabolismo , Antígenos CD19/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Western Blotting , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Citotoxicidade Imunológica/imunologia , Citometria de Fluxo , Células HEK293 , Humanos , Cinética , Leucemia/imunologia , Leucemia/patologia , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Ligação Proteica/imunologia , Estabilidade Proteica , Receptores de IgG/imunologia , Receptores de IgG/metabolismo , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/metabolismo , Temperatura
16.
Mol Cancer ; 9: 301, 2010 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-21092273

RESUMO

BACKGROUND: Advanced melanoma is characterized by a pronounced resistance to therapy leading to a limited patient survival of ~6 - 9 months. Here, we report on a novel bifunctional therapeutic fusion protein, designated anti-MCSP:TRAIL, that is comprised of a melanoma-associated chondroitin sulfate proteoglycan (MCSP)-specific antibody fragment (scFv) fused to soluble human TRAIL. MCSP is a well-established target for melanoma immunotherapy and has recently been shown to provide important tumorigenic signals to melanoma cells. TRAIL is a highly promising tumoricidal cytokine with no or minimal toxicity towards normal cells. Anti-MCSP:TRAIL was designed to 1. selectively accrete at the cell surface of MCSP-positive melanoma cells and inhibit MCSP tumorigenic signaling and 2. activate apoptotic TRAIL-signaling. RESULTS: Treatment of a panel of MCSP-positive melanoma cell lines with anti-MCSP:TRAIL induced TRAIL-mediated apoptotic cell death within 16 h. Of note, treatment with anti-MCSP:sTRAIL was also characterized by a rapid dephosphorylation of key proteins, such as FAK, implicated in MCSP-mediated malignant behavior. Importantly, anti-MCSP:TRAIL treatment already inhibited anchorage-independent growth by 50% at low picomolar concentrations, whereas > 100 fold higher concentrations of non-targeted TRAIL failed to reduce colony formation. Daily i.v. treatment with a low dose of anti-MCSP:TRAIL (0.14 mg/kg) resulted in a significant growth retardation of established A375 M xenografts. Anti-MCSP:TRAIL activity was further synergized by co-treatment with rimcazole, a σ-ligand currently in clinical trials for the treatment of various cancers. CONCLUSIONS: Anti-MCSP:TRAIL has promising pre-clinical anti-melanoma activity that appears to result from combined inhibition of tumorigenic MCSP-signaling and concordant activation of TRAIL-apoptotic signaling. Anti-MCSP:TRAIL alone, or in combination with rimcazole, may be of potential value for the treatment of malignant melanoma.


Assuntos
Proteoglicanas de Sulfatos de Condroitina/metabolismo , Melanoma/tratamento farmacológico , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Proteoglicanas de Sulfatos de Condroitina/genética , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Masculino , Melanócitos/efeitos dos fármacos , Melanócitos/metabolismo , Melanoma/metabolismo , Camundongos , Camundongos Nus , Proteoma/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Br J Haematol ; 150(5): 574-86, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20636437

RESUMO

Two trivalent constructs consisting of single-chain Fv antibody fragments (scFvs) specific for the interleukin-3 receptor alpha chain (CD123), CD33 and the Fcgamma-receptor III (CD16) were designed and characterized for the elimination of acute myeloid leukaemia (AML) cells. The dual targeting single-chain Fv triplebody (sctb) [123 x ds16 x 33] and the mono targeting sctb [123 x ds16 x 123] both specifically bound their respective target antigens and were stable in human serum at 37 degrees C for at least 5 d. Both constructs induced potent antibody-dependent cellular cytotoxicity (ADCC) of two different AML-derived CD33- and CD123 double-positive cell lines in the low picomolar range using isolated mononuclear cells (MNCs) as effector cells. In these experiments the dual targeting molecule produced significantly stronger lysis than the mono targeting agent. In addition, the sctbs showed a high potency in mediating ADCC of primary leukaemia cells isolated from peripheral blood or bone marrow of seven AML patients. Hence, these novel molecules displayed potent anti-leukaemic effects against AML cells in vitro and represent attractive candidates for further preclinical development.


Assuntos
Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Subunidade alfa de Receptor de Interleucina-3/imunologia , Leucemia Mieloide Aguda/imunologia , Anticorpos de Cadeia Única/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Proteínas Ligadas por GPI , Humanos , Fragmentos de Imunoglobulinas/imunologia , Células Matadoras Naturais/imunologia , Leucemia Mieloide Aguda/patologia , Receptores de IgG/imunologia , Proteínas Recombinantes/imunologia , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico , Células Tumorais Cultivadas
18.
Blood ; 116(13): 2277-85, 2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20566893

RESUMO

The use of dendritic cells (DCs) in therapeutic cancer vaccination requires their loading with tumor-specific antigen(s). DEC-205, a phagocytosis receptor mediating antigen uptake, is associated with CD8(+) T-cell responses in mice. Here we fused an anti-DEC-205scFv to an HLA-DP4-restricted epitope from the tumor antigen MAGE-A3, and examined the suitability and efficacy of DEC-205 to deliver a helper epitope to human monocyte-derived DCs (moDCs). The construct specifically bound DEC-205 on human moDCs without negative impact on DC phenotype and function. We measured antigen presentation with specific autologous CD4(+) T cells, generated by TCR-RNA transfection. DEC-205 targeting resulted in significant major histocompatibility complex class II-restricted antigen presentation, and was superior to loading DCs by electroporation of mRNA encoding endosome-targeted MAGE-A3-DCLAMP or by direct peptide pulsing. Anti-DEC-205scFv-MAGE-A3 was presented 100 times more efficiently than the control constructs. DC maturation before or during incubation with anti-DEC-205scFv-MAGE-A3 reduced the interleukin-10/interleukin-2 ratio. Moreover, we successfully applied the DEC-205 targeting strategy to moDCs from malignant melanoma patients. Again, DEC-205-targeted mature DCs (mDCs) presented the antigen more efficiently than peptide-pulsed DCs and maintained their stimulatory capacity after cryoconservation. Thus, DEC-205 targeting represents a feasible and effective method to deliver helper epitopes to DCs in anticancer vaccine strategies, which may also be suitable for DC targeting in vivo.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos CD/imunologia , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Lectinas Tipo C/imunologia , Receptores de Superfície Celular/imunologia , Sequência de Aminoácidos , Anticorpos/administração & dosagem , Anticorpos/genética , Antígenos CD/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/imunologia , Citocinas/biossíntese , Células Dendríticas/citologia , Epitopos/administração & dosagem , Epitopos/genética , Antígenos HLA-DP/metabolismo , Cadeias beta de HLA-DP , Humanos , Imunoterapia Ativa , Técnicas In Vitro , Lectinas Tipo C/metabolismo , Melanoma/imunologia , Melanoma/terapia , Antígenos de Histocompatibilidade Menor , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia
19.
J Immunother ; 33(6): 599-608, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20551837

RESUMO

Single-chain Fv triplebodies (sctb), consisting of a single polypeptide chain with 3 single-chain antibody variable fragments connected in tandem, were generated as antileukemic agents. A CD19-specific sctb of this format has previously been shown to be superior to a bispecific single-chain Fv antibody fragment (bsscFv) for the elimination of leukemic B-lineage cells, but corresponding targeted agents for the treatment of acute myeloid leukemia are still lacking. For this purpose, both a bsscFv and a sctb specific for CD33 and the trigger molecule CD16 (FcgammaRIII) were produced. The sctb displayed 3.5-fold greater avidity for CD33 than the bsscFv 33xds16, whereas both had close to equal affinity for CD16. In antibody-dependent cellular cytotoxicity (ADCC) reactions with human mononuclear cells as effectors, both the bsscFv 33xds16 and the sctb induced lysis of tumor cells with half maximum effective concentrations (EC50) in the low picomolar range. It is interesting to note that the sctb promoted equal lysis of human leukemia-derived cell lines at 10 to 200-fold lower concentrations than the bsscFv. Both molecules mediated ADCC of primary patient cells. In conclusion, both the bsscFv 33xds16 and the sctb 33xds16x33 eliminated acute myeloid leukemia cells in ADCC reactions, but the novel sctb format showed significantly greater specific activity.


Assuntos
Anticorpos Biespecíficos/farmacologia , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Imunoterapia , Leucemia Mieloide Aguda/imunologia , Receptores de IgG/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Animais , Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/genética , Afinidade de Anticorpos , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígenos CD/biossíntese , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/biossíntese , Antígenos de Diferenciação Mielomonocítica/genética , Células CHO , Simulação por Computador , Cricetinae , Cricetulus , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Engenharia de Proteínas , Estabilidade Proteica , Receptores de IgG/genética , Receptores de IgG/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico
20.
J Control Release ; 144(2): 251-8, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20184933

RESUMO

SiRNA molecules represent promising therapeutic molecules, e.g. for cancer therapy. However, efficient delivery into tumor cells remains a major obstacle for treatment. Here, we describe a liposomal siRNA carrier system for targeted delivery of siRNA to CD33-positive acute myeloid leukemia cells. The siRNA is directed against the t(8;21) translocation resulting in the AML1/MTG8 fusion protein. The siRNA was encapsulated in free or polyethylene imine (PEI)-complexed form into PEGylated liposomes endowed subsequently with an anti-CD33 single-chain Fv fragment (scFv) for targeted delivery. The resulting siRNA-loaded immunoliposomes (IL) and immunolipoplexes (ILP) showed specific binding and internalization by CD33-expressing myeloid leukemia cell lines (SKNO-1, Kasumi-1). Targeted delivery of AML1/MTG8 siRNA, but not of mismatch control siRNA, reduced AML1/MTG8 mRNA and protein levels and decreased leukemic clonogenicity, a hallmark of leukemic self-renewal. Although this study revealed that further modifications are necessary to increase efficacy of siRNA delivery and silencing, we were able to establish a targeted liposomal siRNA delivery system combining recombinant antibody fragments for targeted delivery with tumor cell-specific siRNA molecules as therapeutic agents.


Assuntos
Leucemia Mieloide Aguda/genética , Neoplasias/terapia , RNA Interferente Pequeno/genética , Antígenos CD , Antígenos de Diferenciação Mielomonocítica , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/patologia , Lipossomos/administração & dosagem , Neoplasias/genética , Proteínas Proto-Oncogênicas , Proteína 1 Parceira de Translocação de RUNX1 , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico , Fatores de Transcrição , Translocação Genética/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA