Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Regul Toxicol Pharmacol ; 122: 104890, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33587934

RESUMO

Antibody Drug Conjugates (ADCs) are complex multi-domain biotherapeutics which combine, with the aid of a chemical linker, tumor-targeting antibodies with potent small molecule cytotoxicants (also called warhead or payload) for the treatment of cancer. ADCs are a rapidly growing class of pharmaceuticals with nine FDA-approved drugs already on the market and over eighty at different stages of clinical development, and also an increasing number under evaluation for non-oncological indications. Off-target toxicity and a narrow therapeutic index has been a problem with ADCs. This has driven the search for better targeting (disease models, cell surface antigens), linker stability, and payload specificity. Analysis of regulatory approval documents, scientific publications and ICH guidance shows that safety evaluation of ADCs requires novel integrated strategies different from both standard chemotherapy and antibody-based products, e.g. development and validation of ADC analytical assays. There is no ADC-specific guidance on safety evaluation; current guidance emphasises the need for an adaptive approach but more ADC-specific guidance is now arguably possible. The data now available will help to optimize primary target specificity, select appropriate combination partners, develop in silico models, and provide guidance for preclinical and clinical safety evaluation for the next generation of this class of multi-domain therapeutics.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Imunoconjugados/efeitos adversos , Avaliação Pré-Clínica de Medicamentos/normas , Imunoconjugados/farmacocinética , Neoplasias/tratamento farmacológico , Medição de Risco
2.
Rheumatology (Oxford) ; 57(4): 748-755, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29365185

RESUMO

Objectives: We have previously reported that F8-IL4, a fusion protein consisting of the F8 antibody specific to the alternatively-spliced extra domain A of fibronectin and of murine IL-4, cures mice with established arthritis, when used in combination with dexamethasone (DXM). The goal of this study was to assess whether other therapeutic agents, besides DXM, could induce cures in combination with F8-IL4 and to elucidate which leucocytes are most affected by the pharmacological treatment. Methods: We performed therapy experiments in mice with CIA, using intravenous administrations of F8-IL4 in combination with DXM, MTX, murine cytotoxic T-lymphocyte-associated protein 4 fused to the fragment crystallizable portion of murine IgG2a, as well as mAbs to murine IL17A or the p40 subunit of murine IL12/IL23. Histology and immunohistochemistry for the identification of the various leucocytes were performed on the paws of mice euthanized at different therapy time points. Results: Only the use of F8-IL4 in combination with DXM induced complete remissions, while all other combinations did not lead to cures. The light microscopical evaluation of paws with arthritis revealed a predominant infiltration of neutrophils, which substantially decreased 24 h after treatment with F8-IL4 and DXM. Conclusion: The combination of F8-IL4 with DXM promotes a rapid anti-arthritic action by potently inhibiting neutrophil activity. A fully human analogue of F8-IL4 may find clinical utility for the treatment of neutrophil-driven chronic inflammatory conditions.


Assuntos
Artrite/tratamento farmacológico , Proteínas de Transporte/intoxicação , Dexametasona/administração & dosagem , Inflamação/tratamento farmacológico , Interleucina-4/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Artrite/metabolismo , Artrite/patologia , Linhagem Celular , Cricetinae , Modelos Animais de Doenças , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Glucocorticoides/administração & dosagem , Imuno-Histoquímica , Inflamação/metabolismo , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Endogâmicos DBA , Indução de Remissão/métodos
3.
Eur J Immunol ; 46(5): 1246-57, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26919786

RESUMO

F8-IL-4 is a recently developed immunocytokine that delivers IL-4 to sites of inflammation by targeting the neovasculature. We previously reported that F8-IL-4, in combination with dexamethasone (DXM), provides a durable therapy in mice with collagen-induced arthritis (CIA). Therefore, the objective of this study was to identify the mechanism by which IL-4 and DXM combination therapy provides long-lasting disease remission. F8-IL-4 alone attenuated inflammation in CIA and this was associated with increased TH 2 and decreased TH 17 cell numbers in the joints. Similarly, DXM alone had an antiinflammatory effect associated with lower TH 17 cell numbers. In both cases, these therapeutic benefits were reversed once treatment was stopped. On the other hand, combination therapy with F8-IL-4 plus DXM led to a synergistic increase in the percentage of regulatory T (Treg) cells and antiinflammatory macrophages in the arthritic joint and spleen as well as IL-10 levels in serum and spleen. The net result of this was a more pronounced attenuation of inflammation and, more importantly, protection from arthritis relapse post therapy retraction. In conclusion, F8-IL-4 plus DXM is a durable treatment for arthritis that acts by promoting Treg cells in a synergistic manner, and by producing a sustained increase in antiinflammatory macrophages.


Assuntos
Anti-Inflamatórios/uso terapêutico , Artrite Experimental/tratamento farmacológico , Artrite Experimental/imunologia , Dexametasona/uso terapêutico , Tolerância Imunológica , Interleucina-4/uso terapêutico , Macrófagos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Humanizados , Artrite Experimental/terapia , Sinergismo Farmacológico , Quimioterapia Combinada , Imunoconjugados/uso terapêutico , Interleucina-10/sangue , Interleucina-10/imunologia , Interleucina-4/imunologia , Articulações/imunologia , Articulações/fisiopatologia , Camundongos , Camundongos Endogâmicos DBA , Terapia de Alvo Molecular , Baço/imunologia , Baço/fisiopatologia , Células Th17/imunologia , Células Th2/imunologia
4.
Invest New Drugs ; 33(4): 791-800, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25983040

RESUMO

Phosphatidylserine (PS) and other anionic phospholipids, which become exposed on the surface of proliferating endothelial cells, tumor cells and certain leukocytes, have been used as targets for the development of clinical-stage biopharmaceuticals. One of these products (bavituximab) is currently being investigated in Phase 3 clinical trials. There are conflicting reports on the ability of bavituximab and other antibodies to recognize PS directly or through beta-2 glycoprotein 1, a serum protein that is not highly conserved across species. Here, we report on the generation and characterization of two fully human antibodies directed against phosphatidylserine. One of these antibodies (PS72) bound specifically to phosphatidylserine and to phosphatidic acid, but did not recognize other closely related phospholipids, while the other antibody (PS41) also bound to cardiolipin. Both PS72 and PS41 stained 8/9 experimental tumor models in vitro, but both antibodies failed to exhibit a preferential tumor accumulation in vivo, as revealed by quantitative biodistribution analysis. Our findings indicate that anionic phospholipids are exposed and accessible in most tumor types, but cast doubts about the possibility of efficiently targeting tumors in vivo with PS-specific reagents.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias Experimentais/metabolismo , Fosfolipídeos/metabolismo , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Humanos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Nus , Terapia de Alvo Molecular
5.
Exp Cell Res ; 327(1): 135-45, 2014 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-24925479

RESUMO

There is a considerable interest for the discovery and characterization of tumor-associated antigens, which may facilitate antibody-based pharmacodelivery strategies. Thrombospondin-1 and thrombospondin-2 are homologous secreted proteins, which have previously been reported to be overexpressed during remodeling typical for wound healing and tumor progression and to possibly play a functional role in cell proliferation, migration and apoptosis. To our knowledge, a complete immunohistochemical characterization of thrombospondins levels in normal rodent tissues has not been reported so far. Using antibody phage technology, we have generated and characterized monoclonal antibodies specific to murine thrombospondin-1 and thrombospondin-2, two antigens which share 62% aminoacid identity. An immunofluorescence analysis revealed that both antigens are virtually undetectable in normal mouse tissues, except for a weak staining of heart tissue by antibodies specific to thrombospondin-1. The analysis also showed that thrombospondin-1 was strongly expressed in 5/7 human tumors xenografted in nude mice, while it was only barely detectable in 3/8 murine tumors grafted in immunocompetent mice. By contrast, a high-affinity antibody to thrombospondin-2 revealed a much lower level of expression of this antigen in cancer specimens. Our analysis resolves ambiguities related to conflicting reports on thrombosponding expression in health and disease. Based on our findings, thrombospondin-1 (and not thrombospondin-2) may be considered as a target for antibody-based pharmacodelivery strategies, in consideration of its low expression in normal tissues and its upregulation in cancer.


Assuntos
Anticorpos Monoclonais/imunologia , Expressão Gênica/genética , Expressão Gênica/imunologia , Neoplasias/genética , Neoplasias/imunologia , Trombospondina 1/imunologia , Trombospondinas/imunologia , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Nus
6.
Cancer Immunol Immunother ; 63(9): 901-10, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24893857

RESUMO

Antibody-cytokine fusion proteins ("immunocytokines") represent a promising class of armed antibody products, which allow the selective delivery of potent pro-inflammatory payloads at the tumor site. The antibody-based selective delivery of interleukin-2 (IL2) is particularly attractive for the treatment of metastatic melanoma, an indication for which this cytokine received marketing approval from the US Food and drug administration. We used the K1735M2 immunocompetent syngeneic model of murine melanoma to study the therapeutic activity of F8-IL2, an immunocytokine based on the F8 antibody in diabody format, fused to human IL2. F8-IL2 was shown to selectively localize at the tumor site in vivo, following intravenous administration, and to mediate tumor growth retardation, which was potentiated by the combination with paclitaxel or dacarbazine. Combination treatment led to a substantially more effective tumor growth inhibition, compared to the cytotoxic drugs used as single agents, without additional toxicity. Analysis of the immune infiltrate revealed a significant accumulation of CD4(+) T cells 24 h after the administration of the combination. The fusion proteins F8-IL2 and L19-IL2, specific to the alternatively spliced extra domain A and extra domain B of fibronectin respectively, were also studied in combination with tumor necrosis factor (TNF)-based immunocytokines. The combination treatment was superior to the action of the individual immunocytokines and was able to eradicate neoplastic lesions after a single intratumoral injection, a procedure that is being clinically used for the treatment of Stage IIIC melanoma. Collectively, these data reinforce the rationale for the use of IL2-based immunocytokines in combination with cytotoxic agents or TNF-based immunotherapy for the treatment of melanoma patients.


Assuntos
Anticorpos Monoclonais/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Imunoterapia/métodos , Interleucina-2/farmacologia , Melanoma Experimental/terapia , Proteínas Recombinantes de Fusão/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Linhagem Celular Tumoral , Terapia Combinada , Dacarbazina/administração & dosagem , Sinergismo Farmacológico , Feminino , Interleucina-2/administração & dosagem , Interleucina-2/imunologia , Interleucina-2/farmacocinética , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Paclitaxel/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Distribuição Tecidual , Fator de Necrose Tumoral alfa/imunologia
7.
Angew Chem Int Ed Engl ; 53(16): 4231-5, 2014 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-24623670

RESUMO

Antibody-drug conjugates are a very promising class of new anticancer agents, but the use of small-molecule ligands for the targeted delivery of cytotoxic drugs into solid tumors is less well established. Here, we describe the first small-molecule drug conjugates for the treatment of carbonic anhydrase IX expressing solid tumors. Using ligand-dye conjugates we demonstrate that such molecules can preferentially accumulate inside antigen-positive lesions, have fast targeting kinetics and good tumor-penetrating properties, and are easily accessible by total synthesis. A disulfide-linked drug conjugate with the maytansinoid DM1 as the cytotoxic payload and a derivative of acetazolamide as the targeting ligand exhibited a potent antitumor effect in SKRC52 renal cell carcinoma in vivo. It was furthermore superior to sunitinib and sorafenib, both small-molecule standard-of-care drugs for the treatment of kidney cancer.


Assuntos
Anidrases Carbônicas/química , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/terapia , Animais , Sistemas de Liberação de Medicamentos , Humanos , Camundongos , Estrutura Molecular , Pró-Fármacos
8.
Bioconjug Chem ; 21(2): 302-13, 2010 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-20073477

RESUMO

Synthesis of three new cationic thiol-reactive maleimide-porphyrin derivatives and their use in site-specific conjugation to monoclonal antibodies is reported. The selective reactivity toward thiols is demonstrated using competition experiments, where both thiols and amines are present. This selectivity was used to successfully achieve specific conjugation of two porphyrins to cysteine residues present in the antiangiogenic antibody L19, expressed in small immunoprotein (SIP) format. The effect of length and hydrophilicity of the linkage between porphyrin and maleimide was also investigated, and maximum photocytotoxicity was achieved with the longest and most hydrophilic chain. Immunoreactivity and in vitro photocytotoxicity for these well-characterized porphyrin-antibody conjugates are described.


Assuntos
Inibidores da Angiogênese/química , Anticorpos Monoclonais/química , Porfirinas/química , Aminas/química , Animais , Sítios de Ligação , Células CHO , Cricetinae , Cricetulus , Imunoconjugados/química , Imunoproteínas/química , Maleimidas/química , Oxirredução , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/química , Porfirinas/síntese química , Solubilidade , Especificidade por Substrato , Compostos de Sulfidrila/química , Água/química
9.
Immunotherapy ; 10(3): 177-188, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29370721

RESUMO

AIM: The combination of tumor-targeting IL2- and TNF-based antibody-cytokine fusions has exhibited encouraging results in mouse and men. Here, we studied their combination to assess efficacy and mechanism of action in four different immunocompetent mouse models of cancer. METHODS: Mice receiving a single intratumoral injection of F8-IL2, F8-TNF or the combination were investigated for tumor-infiltrating leukocytes and rechallenged when cured. RESULTS: In three models, a proportion of treated animals could be cured, most probably by infiltrating NK and CD8+ T cells. Most of the cured mice did not acquire protective immunity when rechallenged with the same tumor cell line. CONCLUSION: Immunocompetent mouse tumor models may not be adequate enough to predict the search for more efficacious therapy regimens.


Assuntos
Antineoplásicos/administração & dosagem , Imunoterapia/métodos , Interleucina-2/administração & dosagem , Neoplasias/imunologia , Neoplasias/terapia , Fatores de Necrose Tumoral/administração & dosagem , Animais , Anticorpos Monoclonais/administração & dosagem , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Fibronectinas/genética , Injeções Intralesionais , Interleucina-2/genética , Camundongos , Proteínas Recombinantes de Fusão/administração & dosagem , Resultado do Tratamento , Fatores de Necrose Tumoral/genética
10.
Nat Chem ; 10(4): 441-448, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29556050

RESUMO

In nature, specific antibodies can be generated as a result of an adaptive selection and expansion of lymphocytes with suitable protein binding properties. We attempted to mimic antibody-antigen recognition by displaying multiple chemical diversity elements on a defined macrocyclic scaffold. Encoding of the displayed combinations was achieved using distinctive DNA tags, resulting in a library size of 35,393,112. Specific binders could be isolated against a variety of proteins, including carbonic anhydrase IX, horseradish peroxidase, tankyrase 1, human serum albumin, alpha-1 acid glycoprotein, calmodulin, prostate-specific antigen and tumour necrosis factor. Similar to antibodies, the encoded display of multiple chemical elements on a constant scaffold enabled practical applications, such as fluorescence microscopy procedures or the selective in vivo delivery of payloads to tumours. Furthermore, the versatile structure of the scaffold facilitated the generation of protein-specific chemical probes, as illustrated by photo-crosslinking.


Assuntos
Compostos Macrocíclicos/química , Fator de Necrose Tumoral alfa/análise , Calmodulina/análise , Anidrase Carbônica IX/análise , Anidrase Carbônica IX/metabolismo , Peroxidase do Rábano Silvestre/análise , Peroxidase do Rábano Silvestre/metabolismo , Humanos , Microscopia de Fluorescência , Orosomucoide/análise , Antígeno Prostático Específico/análise , Albumina Sérica Humana/análise , Tanquirases/análise , Tanquirases/metabolismo
11.
Clin Cancer Res ; 24(15): 3656-3667, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29691298

RESUMO

Purpose: Antibody-drug conjugates and small-molecule-drug conjugates have been proposed as alternatives to conventional anticancer cytotoxic agents, with the potential to deliver bioactive payloads to the site of disease, helping spare normal tissues.Experimental Design: Here, we describe a novel small-molecule-drug conjugate, based on a high-affinity ligand specific to carbonic anhydrase IX. The product featured a peptidic linker, suitable for cleavage in the tumor extracellular environment, and monomethyl auristatin E as cytotoxic payload.Results: A potent anticancer activity was observed in nude mice bearing SKRC-52 renal cell carcinoma xenografts, but no durable complete responses could be observed in this model. However, when the product was administered together with L19-IL2 (a clinical-stage fusion protein capable of delivering IL2 to the tumor neovasculature), all treated mice in the combination group could be rendered tumor free, in a process that favored the influx of natural killer cells into the tumor mass. The combination of L19-IL2 and the new small-molecule-drug conjugate also eradicated cancer in 100% of immunocompetent mice, bearing subcutaneously grafted CT26 colorectal cancer cells, which stably expressed carbonic anhydrase IX.Conclusions: These findings may be of clinical significance, because carbonic anhydrase IX is overexpressed in the majority of clear cell renal cell carcinomas and in approximately 30% of colorectal cancers. The targeted delivery of IL2 helps potentiate the action of targeted cytotoxics, leading to cancer eradication in models that cannot be cured by conventional chemotherapy. Clin Cancer Res; 24(15); 3656-67. ©2018 AACR.


Assuntos
Anidrase Carbônica IX/antagonistas & inibidores , Carcinoma de Células Renais/tratamento farmacológico , Neoplasias Colorretais/tratamento farmacológico , Interleucina-2/antagonistas & inibidores , Animais , Antígenos de Neoplasias/imunologia , Anidrase Carbônica IX/imunologia , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Imunoterapia/métodos , Interleucina-2/imunologia , Ligantes , Camundongos , Terapia de Alvo Molecular , Peptídeos/imunologia , Peptídeos/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Mol Cancer Ther ; 16(11): 2442-2451, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28716814

RESUMO

A novel biopharmaceutical, consisting of the F8 mAb (specific to a splice isoform of fibronectin) simultaneously fused to both TNF and IL2, was found to react with the majority of solid tumors and hematologic malignancies in mouse and man, but not with healthy adult tissues. The product selectively localized to neoplastic lesions in vivo, as evidenced by quantitative biodistribution studies using radioiodinated protein preparations. When the potency of the cytokine payloads was matched by a single-point mutation, the resulting fusion protein (IL2-F8-TNFmut) eradicated soft-tissue sarcomas in immunocompetent mice, which did not respond to individual antibody-cytokine fusion proteins or by standard doxorubicin treatment. Durable complete responses were also observed in mice bearing CT26, C1498, and F9 tumors. The simultaneous delivery of multiple proinflammatory payloads to the cancer site conferred protective immunity against subsequent tumor challenges. A fully human homolog of IL2-F8-TNFmut, which retained selectivity similar to its murine counterpart when tested on human material, may open new clinical applications for the immunotherapy of cancer. Mol Cancer Ther; 16(11); 2442-51. ©2017 AACR.


Assuntos
Interleucina-2/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Sarcoma/terapia , Fator de Necrose Tumoral alfa/uso terapêutico , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Linhagem Celular Tumoral , Fibronectinas/genética , Fibronectinas/imunologia , Fibronectinas/uso terapêutico , Humanos , Imunoterapia , Interleucina-2/genética , Interleucina-2/imunologia , Camundongos , Mutação Puntual , Proteínas Recombinantes de Fusão/genética , Sarcoma/genética , Sarcoma/patologia , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/uso terapêutico , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
13.
J Nucl Med ; 57(6): 943-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26912427

RESUMO

UNLABELLED: Small organic ligands, selective for tumor-associated antigens, are increasingly being considered as alternatives to monoclonal antibodies for the targeted delivery of diagnostic and therapeutic payloads such as radionuclides and drugs into neoplastic masses. We have previously described a novel acetazolamide derivative, a carbonic anhydrase ligand with high affinity for the tumor-associated isoform IX (CAIX), which can transport highly potent cytotoxic drugs into CAIX-expressing solid tumors. The aim of the present study was to quantitatively investigate the biodistribution properties of said ligand and understand whether acetazolamide conjugates merit further development as drug carriers and radioimaging agents. METHODS: The conjugate described in this study, consisting of a derivative of acetazolamide, a spacer, and a peptidic (99m)Tc chelator, was labeled using sodium pertechnetate under reducing conditions and injected intravenously into CAIX-expressing SKRC-52 xenograft-bearing mice. Animals were sacrificed, and organ uptake as percentage injected activity of radiolabeled ligand per gram of tissues (%IA/g) was evaluated between 10 min and 24 h. Additionally, postmortem imaging by SPECT was performed. RESULTS: The acetazolamide conjugate described in this study could be labeled to high radiochemical purity (>95%, 2.2-4.5 MBq/nmol). Analysis of organ uptake at various time points revealed that the ligand displayed a maximal tumor accumulation 3 h after intravenous injection (22 %IA/g), with an excellent tumor-to-blood ratio of 70:1 at the same time point. The ligand accumulation in the tumor was more efficient than in any other organ, but a residual uptake in the kidney, lung, and stomach (9, 16, and 10 %IA/g, respectively) was also observed, in line with patterns of carbonic anhydrase isoform expression in those tissues. Interestingly, tumor-to-organ ratios improved on administration of higher doses of radiolabeled ligand, suggesting that certain binding sites in normal organs can be saturated in vivo. CONCLUSION: The (99m)Tc-labeled acetazolamide conjugate exhibits high tumor uptake and favorable tumor-to-kidney ratios of up to 3 that may allow imaging of tumors in the kidney and distant sites at earlier time points than commonly possible with antibody-based products. These data suggest that the described molecule merit further development as a radioimaging agent for CAIX-expressing renal cell carcinoma.


Assuntos
Anidrase Carbônica IX/metabolismo , Carcinoma de Células Renais/metabolismo , Neoplasias Renais/metabolismo , Tecnécio/química , Acetazolamida/química , Acetazolamida/metabolismo , Acetazolamida/farmacocinética , Animais , Carcinoma de Células Renais/diagnóstico por imagem , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Marcação por Isótopo , Neoplasias Renais/diagnóstico por imagem , Ligantes , Camundongos , Distribuição Tecidual , Tomografia Computadorizada de Emissão de Fóton Único
14.
Nat Chem ; 7(3): 241-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25698334

RESUMO

In contrast to standard fragment-based drug discovery approaches, dual-display DNA-encoded chemical libraries have the potential to identify fragment pairs that bind simultaneously and benefit from the chelate effect. However, the technology has been limited by the difficulty in unambiguously decoding the ligand pairs from large combinatorial libraries. Here we report a strategy that overcomes this limitation and enables the efficient identification of ligand pairs that bind to a target protein. Small organic molecules were conjugated to the 5' and 3' ends of complementary DNA strands that contain a unique identifying code. DNA hybridization followed by an inter-strand code-transfer created a stable dual-display DNA-encoded chemical library of 111,100 members. Using this approach we report the discovery of a low micromolar binder to alpha-1-acid glycoprotein and the affinity maturation of a ligand to carbonic anhydrase IX, an established marker of renal cell carcinoma. The newly discovered subnanomolar carbonic anhydrase IX binder dramatically improved tumour targeting performance in vivo.


Assuntos
Descoberta de Drogas , Bibliotecas de Moléculas Pequenas , Animais , Anidrases Carbônicas/química , DNA/química , Peroxidase do Rábano Silvestre/química , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Hibridização de Ácido Nucleico , Orosomucoide/química , Estreptavidina/química
15.
Oncotarget ; 6(5): 3043-54, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25460504

RESUMO

Tyrosine kinase inhibitors, affecting angiogenesis, have shown therapeutic efficacy in renal cell carcinoma (RCC). The increased overall survival is not fully explained by their anti-tumor activity, since these drugs frequently induce disease stabilization rather than regression. RCC patients frequently develop cachectic syndrome. We used the RXF393 human renal carcinoma xenograft that recapitulates the characteristics of the disease, including the growth in the mouse kidney (orthotopic implantation), and the induction of cachexia with subsequent premature death. Sunitinib prevents body weight loss and muscle wasting and significantly improves the survival of RXF393-bearing nude mice. The anti-cachectic effect was not associated to direct anti-tumor activity of the drug. Most relevant is the ability of sunitinib to reverse the cachectic phenotype and rescue the animals from the loss of fat tissue. Body weight loss is prevented also in mice bearing the C26 colon carcinoma, classically reported to induce cachexia in immunocompetent mice. Among the mechanisms, we herein show that sunitinib is able to restrain the overactivation of STAT3 and MuRF-1 pathways, involved in enhanced muscle protein catabolism during cancer cachexia. We suggest that off-target effects of angiogenesis inhibitors targeting STAT3 are worth considering as a therapeutic option for patients who develop cachexia, independently of their anti-tumor activity.


Assuntos
Inibidores da Angiogênese/farmacologia , Caquexia/prevenção & controle , Carcinoma de Células Renais/tratamento farmacológico , Indóis/farmacologia , Neoplasias Renais/tratamento farmacológico , Proteínas Musculares/metabolismo , Músculo Esquelético/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Pirróis/farmacologia , Fator de Transcrição STAT3/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Caquexia/diagnóstico por imagem , Caquexia/metabolismo , Caquexia/patologia , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Nus , Músculo Esquelético/diagnóstico por imagem , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestrutura , Transdução de Sinais/efeitos dos fármacos , Sunitinibe , Fatores de Tempo , Proteínas com Motivo Tripartido , Carga Tumoral/efeitos dos fármacos , Redução de Peso/efeitos dos fármacos , Microtomografia por Raio-X , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Cancer Res ; 74(9): 2569-78, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24520075

RESUMO

It is generally thought that the anticancer efficacy of antibody-drug conjugates (ADC) relies on their internalization by cancer cells. However, recent work on an ADC that targets fibronectin in the tumor microenvironment suggests this may not be necessary. The alternatively spliced extra domains A and B (EDA and EDB) of fibronectin offer appealing targets for ADC development, because the antigen is strongly expressed in many solid human tumors and nearly undetectable in normal tissues except for the female reproductive system. In this study, we describe the properties of a set of ADCs based on an antibody targeting the alternatively spliced EDA of fibronectin coupled to one of a set of potent cytotoxic drugs (DM1 or one of two duocarmycin derivatives). The DM1 conjugate SIP(F8)-SS-DM1 mediated potent antitumor activity in mice bearing DM1-sensitive F9 tumors but not DM1-insensitive CT26 tumors. Quantitative biodistribution studies and microscopic analyses confirmed a preferential accumulation of SIP(F8)-SS-DM1 in the subendothelial extracellular matrix of tumors, similar to the pattern observed for unmodified antibody. Notably, we found that treatments were well tolerated at efficacious doses that were fully curative and compatible with pharmaceutical development. Our findings offer a preclinical proof-of-concept for curative ADC targeting the tumor microenvironment that do not rely upon antigen internalization.


Assuntos
Anticorpos/farmacologia , Antineoplásicos/farmacologia , Maitansina/farmacologia , Animais , Anticorpos/uso terapêutico , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Células-Tronco de Carcinoma Embrionário , Feminino , Humanos , Maitansina/farmacocinética , Maitansina/uso terapêutico , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Nus , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Pharmacol Ther ; 139(3): 405-11, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23711792

RESUMO

Metastatic melanoma is one of the most deadly forms of cancer and is poorly responsive to standard chemotherapeutics, such as Dacarbazine and Paclitaxel. Recently, the advent of Vemurafenib and Ipilimumab has broadened the spectrum of therapeutic options for advanced melanoma patients but the occurrence of resistance and of high-grade toxicities call for better and more effective treatments. This review focuses on approved and experimental therapies for metastatic melanoma. The mechanism of action and the reported efficacy data for small molecule drugs and biologics are discussed, outlining directions for future pharmaceutical research in this field.


Assuntos
Antineoplásicos/uso terapêutico , Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Aprovação de Drogas , Desenho de Fármacos , Resistencia a Medicamentos Antineoplásicos , Humanos , Melanoma/patologia , Metástase Neoplásica , Neoplasias Cutâneas/patologia
18.
PLoS One ; 8(8): e72532, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23991120

RESUMO

A sensitive, simple and reproducible protocol for nanoparticle-assisted laser desorption/ionization mass spectrometry imaging technique is described. The use of commercially available TiO2 nanoparticles abolishes heterogeneous crystallization, matrix background interferences and enhances signal detection, especially in the low mass range. Molecular image normalization was based on internal standard deposition on tissues, allowing direct comparison of drug penetration and distribution between different organs and tissues. The method was applied to analyze the distribution of the anticancer drug paclitaxel, inside normal and neoplastic mouse tissue sections. Spatial resolution was good, with a linear response between different in vivo treatments and molecular imaging intensity using therapeutic drug doses. This technique distinguishes the different intensity of paclitaxel distribution in control organs of mice, such as liver and kidney, in relation to the dose. Animals treated with 30 mg/kg of paclitaxel had half of the concentration of those treated with 60 mg/kg. We investigated the spatial distribution of paclitaxel in human melanoma mouse xenografts, following different dosage schedules and found a more homogeneous drug distribution in tumors of mice given repeated doses (5×8 mg/kg) plus a 60 mg/kg dose than in those assigned only a single 60 mg/kg dose. The protocol can be readily applied to investigate anticancer drug distribution in neoplastic lesions and to develop strategies to optimize and enhance drug penetration through different tumor tissues.


Assuntos
Antineoplásicos Fitogênicos/farmacocinética , Espectrometria de Massas/métodos , Melanoma/metabolismo , Nanopartículas , Paclitaxel/farmacocinética , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Calibragem , Feminino , Humanos , Melanoma/tratamento farmacológico , Camundongos , Camundongos Nus , Paclitaxel/uso terapêutico
19.
Clin Cancer Res ; 18(15): 4092-103, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22693354

RESUMO

PURPOSE: Interleukin-12 (IL12) is a potent proinflammatory cytokine with antitumor activity. Its heterodimeric nature makes it compatible with a large variety of different immunocytokine formats. Here we report the design, production, and characterization of a novel immunocytokine, based on the fusion of the F8 antibody (specific to the alternatively spliced EDA domain of fibronectin, a marker of tumor neovasculature) with IL12 (termed IL12-F8-F8). EXPERIMENTAL DESIGN: We developed a novel immunocytokine based on the sequential fusion of interleukin-12 as a single polypeptide with two F8 antibodies in single-chain Fv (scFv) format. The fusion protein was characterized in vitro, and its targeting performance was assessed in vivo. The immunocytokine antitumor activity was studied as monotherapy as well as in combination therapies in three different murine tumor models. Moreover, depletion experiments and tumor analysis revealed a dominant role of natural killer cells for the mechanism of action. RESULTS: IL12-F8-F8 can be produced in mammalian cells, yielding a product of good pharmaceutical quality, capable of selective localization on the tumor neovasculature in vivo, as judged by quantitative biodistribution analysis with radioiodinated protein preparations. The protein potently inhibited tumor growth in three different immunocompetent syngeneic models of cancer. The treatment was generally well tolerated. Moreover, the IL12-F8-F8 fusion protein could be produced both with murine IL12 (mIL12) and with human IL12 (hIL12). CONCLUSIONS: The potent antitumor activity of mIL12-F8-F8, studied alone or in combination with paclitaxel in different tumor models, paves the way to the clinical development of the fully human immunocytokine.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Interleucina-12/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Proteínas Recombinantes de Fusão/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Modelos Animais de Doenças , Feminino , Humanos , Interferon gama/sangue , Interferon gama/imunologia , Interleucina-12/genética , Interleucina-12/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Neoplasias/irrigação sanguínea , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/imunologia , Paclitaxel/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacocinética , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/metabolismo , Distribuição Tecidual , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/imunologia
20.
Cancer Res ; 72(7): 1814-24, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22392081

RESUMO

The selective delivery of bioactive agents to tumors reduces toxicity and enhances the efficacy of anticancer therapies. In this study, we show that the antibody F8, which recognizes perivascular and stromal EDA-fibronectin (EDA-Fn), when conjugated to interleukin-2 (F8-IL2) can effectively inhibit the growth of EDA-Fn-expressing melanomas in combination with paclitaxel. We obtained curative effects with paclitaxel administered before the immunocytokine. Coadministration of paclitaxel increased the uptake of F8 in xenografted melanomas, enhancing tumor perfusion and permeability. Paclitaxel also boosted the recruitment of F8-IL2-induced natural killer (NK) cells to the tumor, suggesting a host response as part of the observed therapeutic benefit. In support of this likelihood, NK cell depletion impaired the antitumor effect of paclitaxel plus F8-IL2. Importantly, this combination reduced both the tumor burden and the number of pulmonary metastatic nodules. The combination did not cause cumulative toxicity. Together, our findings offer a preclinical proof that by acting on the tumor stroma paclitaxel potentiates the antitumor activity elicited by a targeted delivery of IL2, thereby supporting the use of immunochemotherapy in the treatment of metastatic melanoma.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Fitogênicos/uso terapêutico , Fibronectinas/análise , Interleucina-2/uso terapêutico , Melanoma Experimental/tratamento farmacológico , Paclitaxel/uso terapêutico , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Permeabilidade Capilar , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Humanos , Interleucina-2/administração & dosagem , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/química , Melanoma Experimental/patologia , Camundongos , Paclitaxel/administração & dosagem , Isoformas de Proteínas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa