Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 558
Filtrar
1.
Life Sci ; 265: 118791, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33220288

RESUMO

Colorectal cancer (CRC) is the fourth most common cause of cancer and mortality worldwide and is the third most common cancer in men and women. Surgery, radiotherapy, and chemotherapy are conventionally used for the treatment of colorectal cancer. However, these methods are associated with various side effects on normal cells. Thus, new studies are moving towards more effective and non-invasive methods for treatment of colorectal cancer. Targeted therapy of CRC is a promising new approach to enhance the efficiency and decrease the toxicity of the treatment. In targeted therapy of CRC, antibody fragments can directly inhibit tumor cell growth and proliferation. They also can act as an ideal carrier for targeted delivery of anticancer drugs. In the present study, the structure and function of different formats of antibody fragments, immune-targeted therapy of CRC using antibody fragments will be discussed.


Assuntos
Anticorpos Antineoplásicos/administração & dosagem , Anticorpos Antineoplásicos/imunologia , Antineoplásicos/administração & dosagem , Antineoplásicos/imunologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/imunologia , Animais , Biomarcadores Tumorais/imunologia , Desenvolvimento de Medicamentos/tendências , Humanos , Imunoterapia/métodos , Imunoterapia/tendências
2.
Signal Transduct Target Ther ; 5(1): 289, 2020 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-33376237

RESUMO

Advanced natural killer/T cell lymphoma (NKTL) has demonstrated poor prognosis with currently available therapies. Here, we report the efficacy of anti-programmed death 1 (PD-1) antibody with the P-GEMOX (pegaspargase, gemcitabine, and oxaliplatin) regimen in advanced NKTL. Nine patients underwent six 21-day cycles of anti-PD-1 antibody (day 1), pegaspargase 2000 U/m2 (day 1), gemcitabine 1 g/m2 (days 1 and 8) and oxaliplatin 130 mg/m2 (day 1), followed by anti-PD-1 antibody maintenance every 3 weeks. Programmed death-ligand 1 (PD-L1) expression and genetic alterations were determined in paraffin-embedded pretreatment tissue samples using immunohistochemistry and next-generation sequencing (NGS) analysis. Responses were assessed using 18F-fluorodeoxyglucose positron emission tomography (18FDG-PET) and computed tomography or magnetic resonance imaging. Eight patients exhibited significant responses, comprising of seven complete remissions and one partial remission (overall response rate: 88.9%). After a median follow-up of 10.6 months, 6/9 patients (66.7%) remained in complete remission. The most common grade 3/4 adverse events were anemia (33.3%), neutropenia (33.3%), and thrombocytopenia (33.3%); all of which were manageable and resolved. Immunochemotherapy produced a high response rate in patients with positive PD-L1 expression (5/6, 83.3%). NGS analysis suggested that STAT3/JAK3/PD-L1 alterations and ARID1A mutation were associated with immunochemotherapy efficacy. Mutation in DDX3X and alteration in epigenetic modifiers of KMT2D, TET2, and BCORL1 might indicate a poor response to immunochemotherapy. In conclusion, the anti-PD-1 antibody plus P-GEMOX regimen demonstrated promising efficacy in advanced NKTL. PD-L1 expression combined with specific genetic alterations could be used as potential biomarkers to predict therapeutic responses to immunochemotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Fluordesoxiglucose F18/administração & dosagem , Linfoma Extranodal de Células T-NK , Proteínas de Neoplasias/antagonistas & inibidores , Tomografia por Emissão de Pósitrons , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Adulto , Idoso , Anticorpos Antineoplásicos/administração & dosagem , Asparaginase/administração & dosagem , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Intervalo Livre de Doença , Feminino , Seguimentos , Humanos , Linfoma Extranodal de Células T-NK/diagnóstico por imagem , Linfoma Extranodal de Células T-NK/tratamento farmacológico , Linfoma Extranodal de Células T-NK/genética , Linfoma Extranodal de Células T-NK/mortalidade , Masculino , Pessoa de Meia-Idade , Oxaliplatina/administração & dosagem , Polietilenoglicóis/administração & dosagem , Taxa de Sobrevida , Gencitabina
3.
Sci Rep ; 10(1): 20777, 2020 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-33247180

RESUMO

Radionuclide molecular imaging of cancer-specific targets is a promising method to identify patients for targeted antibody therapy. Radiolabeled full-length antibodies however suffer from slow clearance, resulting in high background radiation. To overcome this problem, a pretargeting system based on complementary peptide nucleic acid (PNA) probes has been investigated. The pretargeting relies on sequential injections of primary, PNA-tagged antibody and secondary, radiolabeled PNA probe, which are separated in time, to allow for clearance of non-bound primary agent. We now suggest to include a clearing agent (CA), designed for removal of primary tumor-targeting agent from the blood. The CA is based on the antibody cetuximab, which was conjugated to PNA and lactosaminated by reductive amination to improve hepatic clearance. The CA was evaluated in combination with PNA-labelled trastuzumab, T-ZHP1, for radionuclide HER2 pretargeting. Biodistribution studies in normal mice demonstrated that the CA cleared ca. 7 times more rapidly from blood than unmodified cetuximab. Injection of the CA 6 h post injection of the radiolabeled primary agent [131I]I-T-ZHP1 gave a moderate reduction of the radioactivity concentration in the blood after 1 h from 8.5 ± 1.8 to 6.0 ± 0.4%ID/g. These proof-of-principle results could guide future development of a more efficient CA.


Assuntos
Anticorpos Antineoplásicos/administração & dosagem , Anticorpos Antineoplásicos/química , Imunoconjugados/administração & dosagem , Imunoconjugados/química , Ácidos Nucleicos Peptídicos/administração & dosagem , Ácidos Nucleicos Peptídicos/química , Radioimunoterapia/métodos , Animais , Anticorpos Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Cetuximab/administração & dosagem , Cetuximab/sangue , Cetuximab/química , Feminino , Humanos , Imunoconjugados/farmacocinética , Camundongos , Sondas Moleculares/administração & dosagem , Sondas Moleculares/química , Sondas Moleculares/farmacocinética , Terapia de Alvo Molecular/métodos , Ácidos Nucleicos Peptídicos/farmacocinética , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/imunologia , Distribuição Tecidual , Trastuzumab/administração & dosagem , Trastuzumab/sangue , Trastuzumab/química
4.
Anal Cell Pathol (Amst) ; 2020: 9582731, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32802733

RESUMO

Nanosecond pulsed electric field (NsPEF) ablation effectively eliminates early-stage hepatocellular carcinoma (HCC) by local ablation and advanced HCC by inducing a remarkable and sustained host immune response. However, this approach is not sufficient to prevent cancer progression, and complementary approaches are necessary for effective immunotherapy. In this study, we evaluated the immunoactivating effects and mechanisms of action of nsPEF ablation and PD-1 blockade on an HCC orthotopic xenograft mouse model. Briefly, 24 C57BL-6J tumor-bearing mice were randomly assigned to three groups: nsPEF ablation group, anti-PD-1 administration group, and untreated control group. Tumor-infiltrating T, B, and NK cell levels and plasma concentrations of Th1 (IL-2, IFN-γ, and TNF-α), Th2 (IL-4, IL-5, IL-6, and IL-10), Th9 (IL-9), and Th17 (IL-17A, IL-17F, IL-21, and IL-22) cytokines were evaluated. Both nsPEF ablation and anti-PD-1 treatment induced immune cell infiltration in local tumors and modulated cytokine levels in the peripheral blood, with distinct changes in the two treatment groups. Based on these findings, both nsPEF ablation and PD-1 antibody administration can trigger a local and systemic immune response in a partially complementary manner, and nsPEF ablation should be considered along with PD-1 blockade for the treatment of HCC.


Assuntos
Carcinoma Hepatocelular/imunologia , Eletricidade , Neoplasias Hepáticas/imunologia , Nanopartículas/química , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Anticorpos Antineoplásicos/administração & dosagem , Anticorpos Antineoplásicos/farmacologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Carcinoma Hepatocelular/sangue , Linhagem Celular Tumoral , Citocinas/sangue , Humanos , Imunidade/efeitos dos fármacos , Células Matadoras Naturais/efeitos dos fármacos , Neoplasias Hepáticas/sangue , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Fatores de Tempo
5.
J Surg Res ; 252: 16-21, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32217350

RESUMO

BACKGROUND: Tumor-associated glycoprotein (TAG)-72 is a pancarcinoma antigen that is overexpressed in greater than 80% of colorectal adenocarcinomas. CC49 is a TAG-72-specific antibody. The aim of the present study was to demonstrate selective imaging of colon tumors and metastases with the humanized TAG-72 antibody (anti-huCC49) conjugated to a near-infrared fluorophore in orthotopic mouse models. METHODS: Anti-huCC49 was conjugated to near-infrared dye IR800CW. Mouse imaging was performed with the Pearl Trilogy Small Animal and FLARE Imaging Systems. Subcutaneous mouse models of colon cancer cell line LS174T were used to determine the optimal dose of administration and timing of imaging. Orthotopic mouse models of LS174T were established by surgical orthotopic implantation of LS174T tumors onto the serosa of the cecum. Peritoneal carcinomatosis models were established by injection of LS174T cells into the peritoneum of nude mice. Mice were administered anti-huCC49-IR800 via tail vein injection. Mice were euthanized 72 h later and imaged after laparotomy. RESULTS: Subcutaneous LS174T xenografts demonstrated optimal tumor detection 72 h after administration with 50 µg anti-huCC49-IR800CW. Tumors were visualized with fluorescence imaging with a mean tumor-to-liver ratio of 7.39 (standard deviation: 2.76). In the orthotopic model, metastases smaller than 1 mm were fluorescently visualized that were invisible with bright light. CONCLUSIONS: Anti-huCC49-IR800CW provides sensitive and specific imaging of colon cancer and metastases at a submillimeter resolution in metastatic nude mice models. This provides a promising near-infrared probe for the imaging of colon cancer and metastases for preoperative diagnosis and fluorescence-guided surgery.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Antineoplásicos/administração & dosagem , Antígenos de Neoplasias/imunologia , Neoplasias do Colo/diagnóstico por imagem , Neoplasias Peritoneais/diagnóstico por imagem , Ácidos Alcanossulfônicos/administração & dosagem , Ácidos Alcanossulfônicos/química , Animais , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Antineoplásicos/química , Anticorpos Antineoplásicos/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Neoplasias do Colo/cirurgia , Corantes Fluorescentes/administração & dosagem , Corantes Fluorescentes/química , Humanos , Imunoconjugados/administração & dosagem , Imunoconjugados/química , Imunoconjugados/imunologia , Indóis/administração & dosagem , Indóis/química , Camundongos , Neoplasias Peritoneais/imunologia , Neoplasias Peritoneais/secundário , Neoplasias Peritoneais/cirurgia , Cuidados Pré-Operatórios/métodos , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Cirurgia Assistida por Computador/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Vis Exp ; (143)2019 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-30735151

RESUMO

Potentiation of hostile monoclonal antibodies (mAb) by chemotherapeutic agents constitutes a valuable strategy for designing effective and safer therapy against cancer. Here we provide a protocol to identify a rational combination at the preclinical step. First, we describe a cell-based assay to assess the synergism between anticancer mAb and cytotoxic drugs, that uses the combination index equation of Chou and Talalay1. This includes the measurement of tumor cell drug- and antibody-sensitivity using an MTT assay, followed by an automated computer analysis to calculate the combination index (CI) values. CI values of <1 indicate synergism between tested mAbs and cytotoxic agents1. To corroborate the in vitro findings in vivo, we further describe a method to assess the combination regimen efficacy in a xenograft tumor model. In this model, the combined regimen significantly delays tumor growth, which results in a significant extended survival in comparison to single-agent controls. Importantly, the in vivo experimentation reveals that the combination regimen is well tolerated. This protocol allows the effective evaluation of anticancer drug combinations in preclinical models and the identification of rational combination to evaluate in clinical trials.


Assuntos
Anticorpos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Anticorpos Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Linhagem Celular Tumoral , Interações Medicamentosas , Sinergismo Farmacológico , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Topotecan/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Theranostics ; 8(18): 5106-5125, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30429889

RESUMO

In recent reports, we have shown that optimized pretargeted radioimmunotherapy (PRIT) based on molecularly engineered antibody conjugates and 177Lu-DOTA chelate (DOTA-PRIT) can be used to cure mice bearing human solid tumor xenografts using antitumor antibodies to minimally internalizing membrane antigens, GPA33 (colon) and GD2 (neuroblastoma). However, many solid tumor membrane antigens are internalized after antibody binding and it is generally believed that internalizing tumor membrane antigens are not suitable targets for PRIT. In this study, we tested the hypothesis that DOTA-PRIT can be performed successfully to target HER2, an internalizing membrane antigen widely expressed in breast, ovarian, and gastroesophageal junction cancers. Methods: DOTA-PRIT was carried out in athymic nude mice bearing BT-474 xenografts, a HER2-expressing human breast cancer, using a three-step dosing regimen consisting of sequential intravenous administrations of: 1) a bispecific IgG-scFv (210 kD) format (BsAb) carrying the IgG sequence of the anti-HER2 antibody trastuzumab and the scFv "C825" with high-affinity, hapten-binding antibody for Bn-DOTA (metal) (BsAb: anti-HER2-C825), 2) a 500 kD dextran-based clearing agent, followed by 3) 177Lu-DOTA-Bn. At the time of treatment, athymic nude mice bearing established subcutaneous BT-474 tumors (medium- and smaller-sized tumors with tumor volumes of 209 ± 101 mm3 and ranging from palpable to 30 mm3, respectively), were studied along with controls. We studied single- and multi-dose regimens. For groups receiving fractionated treatment, we verified quantitative tumor targeting during each treatment cycle using non-invasive imaging with single-photon emission computed tomography/computed tomography (SPECT/CT). Results: We achieved high therapeutic indices (TI, the ratio of radiation-absorbed dose in tumor to radiation-absorbed dose to critical organs, such as bone marrow) for targeting in blood (TI = 28) and kidney (TI = 7), while delivering average radiation-absorbed doses of 39.9 cGy/MBq to tumor. Based on dosimetry estimates, we implemented a curative fractionated therapeutic regimen for medium-sized tumors that would deliver approximately 70 Gy to tumors, which required treatment with a total of 167 MBq 177Lu-DOTA-Bn/mouse (estimated absorbed tumor dose: 66 Gy). This regimen was well tolerated and achieved 100% complete responses (CRs; defined herein as tumor volume equal to or smaller than 4.2 mm3), including 62.5% histologic cure (5/8) and 37.5% microscopic residual disease (3/8) at 85 days (d). Treatment controls showed tumor progression to 207 ± 201% of pre-treatment volume at 85 d and no CRs. Finally, we show that treatment with this curative 177Lu regimen leads to a very low incidence of histopathologic abnormalities in critical organs such as bone marrow and kidney among survivors compared with non-treated controls. Conclusion: Contrary to popular belief, we demonstrate that DOTA-PRIT can be successfully adapted to an internalizing antigen-antibody system such as HER2, with sufficient TIs and absorbed tumor doses to achieve a high probability of cures of established human breast cancer xenografts while sparing critical organs of significant radiotoxicity.


Assuntos
Anticorpos Antineoplásicos/administração & dosagem , Neoplasias da Mama/terapia , Terapia de Alvo Molecular/métodos , Octreotida/análogos & derivados , Compostos Organometálicos/administração & dosagem , Radioimunoterapia/métodos , Receptor ErbB-2/metabolismo , Nanomedicina Teranóstica/métodos , Animais , Antígenos de Neoplasias/metabolismo , Neoplasias da Mama/patologia , Modelos Animais de Doenças , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Octreotida/administração & dosagem , Resultado do Tratamento
8.
Theranostics ; 6(12): 2267-2277, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27924162

RESUMO

The complementary nature of positron emission tomography (PET) and near-infrared fluorescence (NIRF) imaging makes the development of strategies for the multimodal PET/NIRF imaging of cancer a very enticing prospect. Indeed, in the context of colorectal cancer, a single multimodal PET/NIRF imaging agent could be used to stage the disease, identify candidates for surgical intervention, and facilitate the image-guided resection of the disease. While antibodies have proven to be highly effective vectors for the delivery of radioisotopes and fluorophores to malignant tissues, the use of radioimmunoconjugates labeled with long-lived nuclides such as 89Zr poses two important clinical complications: high radiation doses to the patient and the need for significant lag time between imaging and surgery. In vivo pretargeting strategies that decouple the targeting vector from the radioactivity at the time of injection have the potential to circumvent these issues by facilitating the use of positron-emitting radioisotopes with far shorter half-lives. Here, we report the synthesis, characterization, and in vivo validation of a pretargeted strategy for the multimodal PET and NIRF imaging of colorectal carcinoma. This approach is based on the rapid and bioorthogonal ligation between a trans-cyclooctene- and fluorophore-bearing immunoconjugate of the huA33 antibody (huA33-Dye800-TCO) and a 64Cu-labeled tetrazine radioligand (64Cu-Tz-SarAr). In vivo imaging experiments in mice bearing A33 antigen-expressing SW1222 colorectal cancer xenografts clearly demonstrate that this approach enables the non-invasive visualization of tumors and the image-guided resection of malignant tissue, all at only a fraction of the radiation dose created by a directly labeled radioimmunoconjugate. Additional in vivo experiments in peritoneal and patient-derived xenograft models of colorectal carcinoma reinforce the efficacy of this methodology and underscore its potential as an innovative and useful clinical tool.


Assuntos
Anticorpos Antineoplásicos/administração & dosagem , Carcinoma/diagnóstico por imagem , Neoplasias Colorretais/diagnóstico por imagem , Radioisótopos de Cobre/administração & dosagem , Corantes Fluorescentes/administração & dosagem , Imagem Óptica/métodos , Tomografia por Emissão de Pósitrons/métodos , Animais , Anticorpos Antineoplásicos/química , Radioisótopos de Cobre/química , Modelos Animais de Doenças , Corantes Fluorescentes/química , Xenoenxertos , Camundongos
9.
ACS Nano ; 10(1): 1512-21, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26727423

RESUMO

Early diagnosis of tumor malignancy is crucial for timely cancer treatment aimed at imparting desired clinical outcomes. The traditional fluorescence-based imaging is unfortunately faced with challenges such as low tissue penetration and background autofluorescence. Upconversion (UC)-based bioimaging can overcome these limitations as their excitation occurs at lower frequencies and the emission at higher frequencies. In this study, multifunctional silica-based nanocapsules were synthesized to encapsulate two distinct triplet-triplet annihilation UC chromophore pairs. Each nanocapsule emits different colors, blue or green, following a red light excitation. These nanocapsules were further conjugated with either antibodies or peptides to selectively target breast or colon cancer cells, respectively. Both in vitro and in vivo experimental results herein demonstrate cancer-specific and differential-color imaging from single wavelength excitation as well as far greater accumulation at targeted tumor sites than that due to the enhanced permeability and retention effect. This approach can be used to host a variety of chromophore pairs for various tumor-specific, color-coding scenarios and can be employed for diagnosis of a wide range of cancer types within the heterogeneous tumor microenvironment.


Assuntos
Adenocarcinoma/diagnóstico , Neoplasias da Mama/diagnóstico , Diagnóstico por Imagem/métodos , Nanocápsulas/química , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Anticorpos Antineoplásicos/administração & dosagem , Anticorpos Antineoplásicos/química , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cor , Feminino , Corantes Fluorescentes/administração & dosagem , Corantes Fluorescentes/química , Humanos , Imunoconjugados/administração & dosagem , Imunoconjugados/química , Injeções Subcutâneas , Células MCF-7 , Camundongos , Camundongos SCID , Células NIH 3T3 , Nanocápsulas/ultraestrutura , Transplante de Neoplasias , Peptídeos/administração & dosagem , Peptídeos/química , Permeabilidade , Transplante Heterólogo
10.
Nature ; 521(7550): 99-104, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25924063

RESUMO

Whereas cancers grow within host tissues and evade host immunity through immune-editing and immunosuppression, tumours are rarely transmissible between individuals. Much like transplanted allogeneic organs, allogeneic tumours are reliably rejected by host T cells, even when the tumour and host share the same major histocompatibility complex alleles, the most potent determinants of transplant rejection. How such tumour-eradicating immunity is initiated remains unknown, although elucidating this process could provide the basis for inducing similar responses against naturally arising tumours. Here we find that allogeneic tumour rejection is initiated in mice by naturally occurring tumour-binding IgG antibodies, which enable dendritic cells (DCs) to internalize tumour antigens and subsequently activate tumour-reactive T cells. We exploited this mechanism to treat autologous and autochthonous tumours successfully. Either systemic administration of DCs loaded with allogeneic-IgG-coated tumour cells or intratumoral injection of allogeneic IgG in combination with DC stimuli induced potent T-cell-mediated antitumour immune responses, resulting in tumour eradication in mouse models of melanoma, pancreas, lung and breast cancer. Moreover, this strategy led to eradication of distant tumours and metastases, as well as the injected primary tumours. To assess the clinical relevance of these findings, we studied antibodies and cells from patients with lung cancer. T cells from these patients responded vigorously to autologous tumour antigens after culture with allogeneic-IgG-loaded DCs, recapitulating our findings in mice. These results reveal that tumour-binding allogeneic IgG can induce powerful antitumour immunity that can be exploited for cancer immunotherapy.


Assuntos
Anticorpos Antineoplásicos/imunologia , Antígenos de Neoplasias/imunologia , Células Dendríticas/imunologia , Imunoglobulina G/imunologia , Neoplasias/imunologia , Linfócitos T/imunologia , Animais , Anticorpos Antineoplásicos/administração & dosagem , Antígenos CD40/metabolismo , Modelos Animais de Doenças , Feminino , Imunoglobulina G/administração & dosagem , Isoanticorpos/administração & dosagem , Isoanticorpos/imunologia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Metástase Neoplásica , Transplante de Neoplasias/imunologia , Neoplasias/patologia , Receptores de IgG/imunologia , Fator de Necrose Tumoral alfa/imunologia
11.
Expert Opin Biol Ther ; 15(6): 787-801, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25882106

RESUMO

INTRODUCTION: After years of limited success, progress of anti-cancer immuno-therapeutics has been considerable over the past decade. Key to this progress has been the application of new biological insights around the importance and nature of immune checkpoints that are able to reverse down-regulation of anti-tumor immunity. AREAS COVERED: An overview of the preclinical and recent clinical trial data on key immuno-regulatory agents currently in development, including antibody targeting of cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed death receptor 1 (PD-1) on T-lymphocytes and its principal ligand (PD-L1) on tumor cells as well as immune agonists (e.g., anti-CD40). EXPERT OPINION: Durable long-term responses in some patients with advanced melanoma, initially with ipilimumab (anti-CTLA-4) and more recently antibodies targeting either PD-1 or PD-L1 in patients with melanoma and renal cancer, non-small-cell lung, bladder and head and neck cancers with less toxicity, have provided real optimism that immunotherapeutic approaches can improve outcomes in a wide range of cancer. The manageable tolerability of PD-1-pathway blockers and their unique mechanism of action are encouraging combination approaches. Current efforts focus on registration trials of single agents plus combinations in many different tumor types and treatment settings and identifying and developing predictive biomarkers of immunological response.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antineoplásicos/imunologia , Imunoterapia/tendências , Neoplasias/imunologia , Neoplasias/terapia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Antineoplásicos/administração & dosagem , Antígeno CTLA-4/imunologia , Humanos , Ipilimumab , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T/imunologia , Resultado do Tratamento
12.
Blood ; 124(24): 3553-60, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25301708

RESUMO

CD19 is ubiquitously expressed on chronic lymphocytic leukemia (CLL) cells and is therefore an attractive candidate for antibody targeting. XmAb5574 (aka MOR00208) is a novel humanized CD19 monoclonal antibody with an engineered Fc region to enhance Fcγ receptor binding affinity. Here we report results of a first in human phase 1 trial of XmAb5574 in patients with relapsed or refractory CLL. Twenty-seven patients were enrolled to 6 escalating dose levels, with expansion at the highest dose level of 12 mg/kg. Nine doses of XmAb5574 were infused over 8 weeks. No maximal tolerated dose was reached, and the drug was generally well tolerated, with infusion reactions of grades 1 and 2 being the most common toxicities. Grade 3 and 4 toxicities occurred in 5 patients and included neutropenia, thrombocytopenia, increased aspartate aminotransferase, febrile neutropenia, and tumor lysis syndrome. XmAb5574 showed preliminary efficacy, with 18 patients (66.7%) responding by physical examination criteria and laboratory studies, and 8 patients (29.6%) responding by computed tomography criteria. Pharmacokinetics showed a half-life of 14 days with clearance that was not dose-dependent. In conclusion, this phase 1 trial demonstrates safety and preliminary efficacy of a novel Fc-engineered CD19 monoclonal antibody XmAb5574 and justifies movement into the phase 2 setting. This trial was registered at www.clinicaltrials.gov as #NCT01161511.


Assuntos
Anticorpos Monoclonais , Anticorpos Antineoplásicos/administração & dosagem , Antígenos CD19 , Antineoplásicos , Leucemia Linfocítica Crônica de Células B/sangue , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Feminino , Meia-Vida , Humanos , Regiões Constantes de Imunoglobulina/administração & dosagem , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Recidiva , Fatores de Tempo , Tomografia Computadorizada por Raios X
13.
Int Immunopharmacol ; 18(1): 20-6, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24239629

RESUMO

To construct an improved biological missile, an immunoconjugate ADM-Dex-ScFv-SA3 was synthesized, which was composed of a hepatocellular carcinoma-specific, single-chain Fv antibody (ScFv-SA3) and a highly potent cytotoxic drug, adriamycin (ADM), as the warhead. Oxidized Dextran T10 (Dex-T10) was used as a linker to connect these two moieties. The 40 KD soluble anti-hepatoma human Trx-ScFv-SA3 protein was expressed in E. coli BL21 (DE3), using a prokaryotic expression vector, pET21a (+)-Trx-ScFv-SA3-His. It was purified using a His-Tag Ni-Agarose column and identified by western blot. The activity of Trx-ScFv-SA3 was verified by enzyme-linked immunosorbent assay (ELISA) and immunocytochemistry to confirm that it specifically binds to the hepatocellular carcinoma cell line HepG2. To prepare ADM-Dex-ScFv-SA3, ADM was conjugated to the antibody at a molar ratio of 14.21:1. The antitumor effect of the conjugate was tested by MTT assay, plate colony formation assay and xenografts in a nude mice experimental model. In vitro experiments revealed that ADM-Dex-ScFv-SA3 could bind to tumor cells selectively and inhibit the proliferation and the colony formation ability of HepG2 cells. In vivo experiments showed that ADM-Dex-ScFv-SA3 suppressed the tumor growth and prolonged the median survival time in tumor-bearing mice. Tumor histology slides indicated a significantly slower tumor tissue proliferation in the ADM-Dex-ScFv-SA3 group. These data indicate that the targeted drug, ADM-Dex-ScFv-SA3, may be a highly potent and selective therapy for the treatment of hepatoma.


Assuntos
Anticorpos Antineoplásicos/administração & dosagem , Antineoplásicos/administração & dosagem , Carcinoma Hepatocelular/terapia , Doxorrubicina/administração & dosagem , Hepatócitos/efeitos dos fármacos , Neoplasias Hepáticas Experimentais/terapia , Anticorpos de Cadeia Única/administração & dosagem , Animais , Anticorpos Antineoplásicos/química , Anticorpos Antineoplásicos/genética , Antineoplásicos/química , Carcinoma Hepatocelular/imunologia , Proliferação de Células/efeitos dos fármacos , Doxorrubicina/química , Vetores Genéticos/genética , Células Hep G2 , Hepatócitos/patologia , Humanos , Neoplasias Hepáticas Experimentais/imunologia , Camundongos , Camundongos Nus , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/genética , Carga Tumoral/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Cancer Res ; 73(8): 2619-27, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23396586

RESUMO

Recruitment of immune cells to tumor cells targeted by a therapeutic antibody can heighten the antitumor efficacy of the antibody. For example, p185(her2/neu)-targeting antibodies not only downregulate the p185(her2/neu) kinase (ERBB2) but also trigger complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) through the antibody Fc region. Here, we describe a generalized strategy to improve immune cell recruitment to targeted cancer cells, using a modified scFv antibody we call a "Grababody" that binds the target protein and endogenous immunoglobulins. The model system we used to illustrate the use of this platform recognizes p185(her2/neu) and includes an IgG binding domain. The recombinant scFv Grababody that was created recruited circulating human IgGs and attracted immune cells carrying Fc receptors to tumor cells that expressed p185(her2/neu). The presence of the IgG binding domain significantly enhanced CDC and ADCC activity and improved antitumor activity in vivo. Our results illustrate a novel general approach to improve antibody-like proteins for therapeutic applications.


Assuntos
Anticorpos Antineoplásicos/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Neoplasias/imunologia , Anticorpos de Cadeia Única/imunologia , Animais , Anticorpos Antineoplásicos/administração & dosagem , Anticorpos Antineoplásicos/metabolismo , Afinidade de Anticorpos/imunologia , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Linhagem Celular , Ativação do Complemento , Citotoxicidade Imunológica , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Camundongos , Neoplasias/terapia , Ligação Proteica , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Receptores de IgG/imunologia , Receptores de IgG/metabolismo , Anticorpos de Cadeia Única/administração & dosagem , Anticorpos de Cadeia Única/metabolismo , Transplante Heterólogo
15.
Curr Cancer Drug Targets ; 13(1): 11-29, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23030233

RESUMO

Pulmonary delivery of drug-loaded nanoparticles is a novel approach for lung cancer treatment and the conjugation of nanoparticles to a targeting ligand further promotes specificity of the carrier cargo to cancer cells. Notably, the epithelial cell adhesion molecule (EpCAM, CD326) is over expressed in lung cancer. Here, we report the safety and proof-of-concept efficacy of drug-loaded nanoparticles and EpCAM immunonanoparticles in a c-Raf transgenic lung cancer model. PEG-PLA nanoparticles and immunonanoparticles were prepared whereby paclitaxel palmitate (Pcpl) was incorporated as a medication for its common use in lung cancer treatment. Four doses of aerosolized nanoparticle formulations or vehicle were endotracheally administered to mice by consecutive or alternate regimes. Pulmonary delivery of drug loaded nano- and/or immunonanoparticle formulations elicited mild inflammation as evidenced by the slightly increased neutrophil and activated macrophage counts in bronchoalveolar lavage. No evidence for pulmonary toxicity following treatment with either blank or drug-loaded nano- and/or immunonanoparticles was observed. Proof-of-concept efficacy was determined by serial CT scanning and histopathology. Animals treated with either EpCAM antibody or Pcpl solution or drug loaded nano- or immunonanoparticles inhibited disease progression. Conversely, disease progression was noted with vehicle treated animals with nearly 30% loss of their aerated lung volume. Importantly, treatment of mice with either Pcpl or EpCAM antibody solution caused 80% mortality and/or haemorrhage, respectively, thus causing unacceptable toxicity. In contrast, the survival of animals treated with either nano- or immunonanoparticles was 60 and 70%, respectively. Taken collectively, pulmonary delivered drug-loaded nano- and EpCAM immunonanoparticles were well tolerated and can be considered a promising strategy for improving lung cancer treatment.


Assuntos
Anticorpos Imobilizados/administração & dosagem , Anticorpos Antineoplásicos/administração & dosagem , Antineoplásicos Fitogênicos/administração & dosagem , Moléculas de Adesão Celular/antagonistas & inibidores , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Neoplasias Pulmonares/tratamento farmacológico , Administração por Inalação , Animais , Anticorpos Imobilizados/efeitos adversos , Anticorpos Imobilizados/uso terapêutico , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/efeitos adversos , Anticorpos Antineoplásicos/uso terapêutico , Antígenos de Neoplasias/metabolismo , Antineoplásicos Fitogênicos/efeitos adversos , Antineoplásicos Fitogênicos/uso terapêutico , Moléculas de Adesão Celular/metabolismo , Sistemas de Liberação de Medicamentos/efeitos adversos , Molécula de Adesão da Célula Epitelial , Feminino , Humanos , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Transgênicos , Nanopartículas/administração & dosagem , Nanopartículas/efeitos adversos , Nanopartículas/química , Nanopartículas/uso terapêutico , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos , Paclitaxel/análogos & derivados , Paclitaxel/uso terapêutico , Palmitatos/administração & dosagem , Palmitatos/efeitos adversos , Palmitatos/uso terapêutico , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Distribuição Aleatória , Propriedades de Superfície , Análise de Sobrevida
16.
Rev. cuba. invest. bioméd ; 31(4): 417-428, oct.-dic. 2012.
Artigo em Espanhol | LILACS | ID: lil-660153

RESUMO

La liberación controlada de fármacos en el sitio del tumor y el desarrollo de técnicas no invasivas de monitoreo constituyen 2 de los principales retos que enfrentan las terapias antitumorales en la actualidad. En este trabajo se analizan algunas de las potencialidades del uso de liposomas como vehículos para el transporte de drogas hasta los tumores, especialmente de las variantes direccionalizadas a antígenos tumorales mediante el acoplamiento de anticuerpos (inmunoliposomas). Estas vesículas pueden a su vez ser utilizadas en combinación con el uso de imágenes de resonancia magnética, una de las técnicas de imagenología más utilizadas y de mayores potencialidades en la visualización a nivel molecular. El uso conjunto de estas 2 tecnologías permite controlar la cantidad de fármaco administrado, así como predecir la eficacia del tratamiento y monitorear la progresión de este


Controlled release of drugs at the tumor site and the development of non-invasive monitoring techniques are two of the main challenges currently facing antitumoral therapies. The paper analyzes some of the potential uses of liposomes as vehicles for the transport of drugs to the tumors, particularly directionalized variants to tumor antigens through antibody coupling (immunoliposomes). These vesicles may also be used in combination with magnetic resonance, one of the most widely used imaging techniques, and one exhibiting great visualization potential at molecular level. Joint use of these two techniques makes it possible to control the amount of drug administered, as well as predict the efficacy of the treatment and monitor its progress


Assuntos
Anticorpos Antineoplásicos/administração & dosagem , Imageamento por Ressonância Magnética/métodos , Lipossomos/uso terapêutico , Neoplasias/terapia
17.
MAbs ; 4(2): 256-66, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22453099

RESUMO

The safety and pharmacokinetics assessment of antibodies targeting CD22 (e.g., epratuzumab) have been established in western Caucasian populations, but there are no reports of the effects in Chinese populations. This dose-escalation study examines the safety, pharmacokinetics and biologic effects of multiple doses of anti-CD22 human-murine chimeric monoclonal antibody SM03 in 21 Chinese patients with CD22-positive non-Hodgkin lymphoma. Most of drug-related adverse events (AEs) were mild and reversible. Two patients experienced serious AEs (hemorrhage); one patient had grade 4 neutropenia; one patient had asymptomatic grade III prolongation of activated partial thromboplastin time (APTT). Major AEs included fever (71%), prolongation of APTT (42.8%), leukocytopenia (44.4%), alanine transaminase elevation (28.6%), elevated serum creatinine (23.8%) and injection site skin redness (14.3%). Circulating B cells transiently decreased without significant effects on T cells or immunoglobulin levels. Pharmacokinetic data revealed that mean maximum observed SM03 concentration and mean AUC from time zero to infinity increased in a dose-dependent manner up to 360 mg/m (2) SM03. Mean clearance was similar at doses ≤ 360 mg/m (2) and decreased significantly at dose 480 mg/m (2), supporting saturation of B-cell binding at 360 mg/m (2). Across all dose levels and histologies, one patient achieved partial response at 480 mg/m (2) dose; 14 patients had stable disease as best response and four patients progressed. Overall, SM03 was tolerated at doses ranging from 60-480 mg/m (2) and had potential efficacy in Chinese patients with follicular lymphoma.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Anticorpos Antineoplásicos/administração & dosagem , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Linfoma de Células B/tratamento farmacológico , Linfoma Folicular/tratamento farmacológico , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico , Adulto , Idoso , Animais , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/imunologia , Anticorpos Antineoplásicos/efeitos adversos , Anticorpos Antineoplásicos/imunologia , Antineoplásicos/efeitos adversos , Antineoplásicos/imunologia , China , Relação Dose-Resposta a Droga , Feminino , Humanos , Linfoma de Células B/imunologia , Linfoma de Células B/patologia , Linfoma Folicular/imunologia , Linfoma Folicular/patologia , Masculino , Camundongos , Pessoa de Meia-Idade
18.
Cancer Biother Radiopharm ; 27(1): 36-40, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22239432

RESUMO

Data was analyzed from 92 patients > 5 years after intraperitoneal (IP) radionuclide therapy (RIT) with (90)Y- or (177)Lu-CC49 to determine prognostic factors. Patients had CC49 antibody-reactive ovarian cancer confined to the abdominal cavity after primary debulking and chemotherapy. The first 27 patients received IP (177)Lu-CC49 alone; the remainder received Interferon (IFN), to increase the expression of the tumor-associated glycoprotein-72 (TAG-72) antigen, +/- IP paclitaxel (25-100 mg/m(2)) 2 days before RIT. Factors assessed by univariate (and some multivariate) analysis included age, race, body size, interval between initial diagnosis and RIT, interval between 2nd look surgery and RIT, (90)Y versus (177)Lu, MBq dose, paclitaxel dose, grade of tumor, extent of initial surgery, size of disease deposits prior to RIT, intensity of TAG reactivity, the addition of unlabeled antibody, and the development of human anti-mouse antibody and/or serum sickness after murine antibody. A statistically significant improvement in progression-free survival (p ≤ 0.05) was noted for less bulky disease and younger age. Administration of paclitaxel plus IFN, an immune response, and use of (90)Y showed a favorable nonsignificant trend. Dose escalation of radionuclide did not change risk of progression; thus, this therapy may have therapeutic efficacy at modest dose levels.


Assuntos
Lutécio/administração & dosagem , Neoplasias Ovarianas/radioterapia , Radioimunoterapia/métodos , Compostos Radiofarmacêuticos/administração & dosagem , Radioisótopos de Ítrio/administração & dosagem , Adulto , Idoso , Animais , Anticorpos Antineoplásicos/administração & dosagem , Anticorpos Antineoplásicos/imunologia , Intervalo Livre de Doença , Feminino , Humanos , Imunotoxinas/administração & dosagem , Imunotoxinas/imunologia , Infusões Parenterais , Camundongos , Pessoa de Meia-Idade , Neoplasias Ovarianas/imunologia , Prognóstico , Compostos Radiofarmacêuticos/imunologia
19.
Mol Imaging Biol ; 14(2): 237-44, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21494920

RESUMO

PURPOSE: The objective of this study is to evaluate the therapeutic response to a novel monoclonal antibody targeting human extracellular matrix metalloproteinase inducer (EMMPRIN) in combination with gemcitabine in a pancreatic-tumor xenograft murine model by sequential 2-deoxy-2-[18F]fluoro-D-glucose ((18)F-FDG) positron emission tomography/computed tomgraphy (PET/CT) imaging. PROCEDURES: Four groups of SCID mice bearing orthotopic pancreatic tumor xenografts were injected with phosphate-buffered saline, gemcitabine (120 mg/kg BW), anti-EMMPRIN antibody (0.2 mg), or combination, respectively, twice weekly for 2 weeks, while (18)F-FDG PET/CT imaging was performed weekly for 3 weeks. Changes in mean standardized uptake value (SUV(mean)) of (18)F-FDG and volume of tumors were determined. RESULTS: The tumor SUV(mean) change in the group receiving combination therapy was significantly lower than those of the other groups. Tumor-volume changes of groups treated with anti-EMMPRIN monotherapy or combined therapy were significantly lower than that of the control group. CONCLUSIONS: These data provide support for clinical studies of anti-EMMPRIN therapy with gemcitabine for pancreatic cancer treatment.


Assuntos
Anticorpos Antineoplásicos/uso terapêutico , Basigina/imunologia , Desoxicitidina/análogos & derivados , Fluordesoxiglucose F18 , Imagem Multimodal , Neoplasias Pancreáticas/tratamento farmacológico , Tomografia por Emissão de Pósitrons , Tomografia Computadorizada por Raios X , Ensaios Antitumorais Modelo de Xenoenxerto , Trifosfato de Adenosina/metabolismo , Animais , Anticorpos Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Meios de Contraste , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Feminino , Humanos , Injeções Intravenosas , Camundongos , Camundongos SCID , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/patologia , Tomografia Computadorizada de Emissão de Fóton Único , Resultado do Tratamento , Gencitabina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA