Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Pharm ; 18(10): 3854-3861, 2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34543035

RESUMO

Tumor necrosis factor-related apoptosis ligand (TRAIL) is a promising protein candidate for selective apoptosis of a variety of cancer cells. However, the short half-life and a lack of targeted delivery are major obstacles for its application in cancer therapy. Here, we propose a simple strategy to solve the targeting problem by genetically fusing an anti-HER2 affibody to the C-terminus of the TRAIL. The fusion protein TRAIL-affibody was produced as a soluble form with high yield in recombinant Escherichia coli. In vitro studies proved that the affibody domain promoted the cellular uptake of the fusion protein in the HER2 overexpressed SKOV-3 cells and improved its apoptosis-inducing ability. In addition, the fusion protein exhibited higher accumulation at the tumor site and greater antitumor effect than those of TRAIL in vivo, indicating that the affibody promoted the tumor homing of the TRAIL and then improved the therapeutic efficacy. Importantly, repeated injection of high-dose TRAIL-affibody showed no obvious toxicity in mice. These results demonstrated that the engineered TRAIL-affibody is promising to be a highly tumor-specific and targeted cancer therapeutic agent.


Assuntos
Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos , Receptor ErbB-2/administração & dosagem , Ligante Indutor de Apoptose Relacionado a TNF/administração & dosagem , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Transplante de Neoplasias , Engenharia de Proteínas/métodos , Receptor ErbB-2/uso terapêutico , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico
2.
Immunopharmacol Immunotoxicol ; 42(4): 346-357, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32515626

RESUMO

BACKGROUND: Human epidermal growth factor receptor 2 (HER2) is overexpressed in a subset of cancers including 25% of breast cancers. Since combination therapy consisting of multiple therapeutic approaches is considered a promising regimen, we examined combination treatment modalities in a xenograft model in Balb/c mice injected with 4T1-HER2 cells. We used HER2/neu-loaded bone marrow-derived dendritic cells (BM-DC's) along with anti-PD-L1 monoclonal antibody in a new combination immunotherapy model. METHODS: The combination was composed of an active immunotherapy (i.e. BM-DC-based vaccine) designed to boost the immune response against target antigen and was augmented by using anti-PD-L1 mAb to prevent immune evasion by the xenografted tumors. The vaccine combination was further supported using a QS-21 saponin adjuvant and the immune response was evaluated. RESULTS: Mice treated with HER2/neu-loaded BM-DCs, combined with QS-21 and anti-PD-L1 mAb had significantly decreased tumor sizes and their splenocytes had enhanced cytotoxic activity, based on the lactate dehydrogenase (LDH) assay, compared to vaccine and adjuvant groups alone. The same vaccination group demonstrated a remarkable increase in IFN-γ secreting CD8+ T-cells analyzed by flow cytometry. ELISA data also revealed a significant increase in the serum anti-HER2 IgG1 response; in addition, there was significant splenocyte proliferation upon stimulation with antigen compared to vaccine and adjuvant groups. Consistently, a significant infiltration of CD4+, CD8+ immune cells in and around the tumors was observed. CONCLUSIONS: Our data suggest that the BM-DC + HER2/neu + QS-21 + anti-PD-L1 vaccine combination paradigm synergistically generates anti-tumor activity and immune responses against HER2 overexpressing breast cancer in mice.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Células Dendríticas/transplante , Fragmentos de Peptídeos/administração & dosagem , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/genética , Saponinas/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Animais , Antineoplásicos Imunológicos/administração & dosagem , Neoplasias da Mama/patologia , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral , Terapia Combinada/métodos , Feminino , Humanos , Imunoterapia Ativa/métodos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
3.
Breast Cancer Res Treat ; 147(2): 401-5, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25129345

RESUMO

The aim of this study is to estimate the frequency of pathologic complete response (pCR) after neoadjuvant treatment with cisplatin chemotherapy in women with breast cancer and a BRCA1 mutation. One hundred and seven women with breast cancer and a BRCA1 mutation, who were diagnosed with stage I to III breast cancer between December 2006 and June 2014, were treated with cisplatin 75 mg/m(2) every 3 weeks for four cycles, followed by mastectomy and conventional chemotherapy. Information was collected on clinical stage, grade, hormone receptor status, and Her2neu status prior to treatment. pCR was determined by review of surgical specimens. One hundred and seven patients were enrolled in the study, including 93 patients who were treated for first primary breast cancer and 14 patients who had previously received treatment for a prior cancer. A pCR was observed in 65 of the 107 patients (61 %). Platinum-based chemotherapy is effective in a high proportion of patients with BRCA1-associated breast cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proteína BRCA1/biossíntese , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Adulto , Idoso , Proteína BRCA1/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/cirurgia , Cisplatino/administração & dosagem , Ciclofosfamida/administração & dosagem , Doxorrubicina/administração & dosagem , Feminino , Humanos , Pessoa de Meia-Idade , Terapia Neoadjuvante , Estadiamento de Neoplasias , Receptor ErbB-2/administração & dosagem , Adulto Jovem
4.
Oncologist ; 18(2): 221-31, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23335619

RESUMO

Molecularly targeted therapeutics (MTT) are the future of cancer systemic therapy. They have already moved from palliative therapy for advanced solid malignancies into the setting of curative-intent treatment for early-stage disease. Cardiotoxicity is a frequent and potentially serious adverse complication of some targeted therapies, leading to a broad range of potentially life-threatening complications, therapy discontinuation, and poor quality of life. Low-cost pleiotropic interventions are therefore urgently required to effectively prevent and/or treat MTT-induced cardiotoxicity. Aerobic exercise therapy has the unique capacity to modulate, without toxicity, multiple gene expression pathways in several organ systems, including a plethora of cardiac-specific molecular and cell-signaling pathways implicated in MTT-induced cardiac toxicity. In this review, we examine the molecular signaling of antiangiogenic and HER2-directed therapies that may underpin cardiac toxicity and the hypothesized molecular mechanisms underlying the cardioprotective properties of aerobic exercise. It is hoped that this knowledge can be used to maximize the benefits of small molecule inhibitors, while minimizing cardiac damage in patients with solid malignancies.


Assuntos
Exercício Físico/fisiologia , Cardiopatias/induzido quimicamente , Cardiopatias/terapia , Neoplasias/terapia , Receptor ErbB-2/uso terapêutico , Inibidores da Angiogênese/efeitos adversos , Inibidores da Angiogênese/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/metabolismo
5.
J Immunol ; 184(8): 4170-7, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20220087

RESUMO

By studying BALB/c mice deficient in immune components, we show that the protective immunity to rat ErbB2(+) tumors rests on the Ab response elicited by the electroporation of a DNA vaccine encoding the extracellular and transmembrane domains of rat ErbB2. In vivo, the adoptive transfer of vaccine-elicited anti-rat ErbB2 Abs protected against a challenge of rat ErbB2(+) carcinoma cells (TUBO cells). In vitro, such Abs inhibited TUBO cell growth by impairing cell cycle progression and inducing apoptosis. To correlate intrinsic mechanisms of Ab action with their tumor-inhibitory potential, first we showed that TUBO cells constitutively express phosphorylated transgenic ErbB2/autochthonous ErbB3 heterodimers and exhibit a basal level of Akt phosphorylation, suggesting a constitutive activation of the PI3K/Akt pathway. Treatment with anti-ErbB2 Abs caused a drastic reduction in the basal level of Akt phosphorylation in the absence of an impairment of PI3K enzymatic activity. Notably, the same Ab treatment induced an increase in PTEN phosphatase activity that correlated with a reduced PTEN phosphorylation. In conclusion, vaccine-induced anti-ErbB2 Abs directly affected the transformed phenotype of rat ErbB2(+) tumors by impairing ErbB2-mediated PI3K/Akt signaling.


Assuntos
Anticorpos Antineoplásicos/fisiologia , Neoplasias Mamárias Experimentais/prevenção & controle , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptor ErbB-2/imunologia , Transdução de Sinais/imunologia , Vacinas de DNA/imunologia , Células 3T3 , Animais , Anticorpos Antineoplásicos/biossíntese , Linhagem Celular Tumoral , Eletroporação , Espaço Extracelular/genética , Espaço Extracelular/imunologia , Feminino , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , PTEN Fosfo-Hidrolase/fisiologia , Fosforilação/imunologia , Estrutura Terciária de Proteína/genética , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/biossíntese , Receptor ErbB-2/genética , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética
6.
J Immunol ; 182(9): 5873-81, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19380836

RESUMO

Modulation of the immune system to amplify anti-tumor immunity carries the risk of developing autoimmune diseases, including hypothyroidism, as seen with cancer patients undergoing clinical trials for immunotherapeutic regimens. Although there is a tendency to view autoimmunity as a positive indicator for cancer immunotherapy, some autoimmune manifestations can be life-threatening and necessitate prolonged medical intervention or removal from trial. We have established murine test models to assess such risks by monitoring, simultaneously, the immune reactivity to tumor-associated rat erbB-2 (neu) and another self Ag, mouse thyroglobulin (mTg). We previously reported that in wild-type, thyroiditis-resistant BALB/c mice that underwent regression of neu(+) TUBO tumors following regulatory T cell (Treg) depletion, immune responses to rat neu and mTg with resultant autoimmune thyroiditis (EAT) were both enhanced. In this study, we tested the balance between tumor immunity and autoimmunity in neu-transgenic BALB NeuT female mice. First, growth and progression of neu(+) tumor were compared in neu tolerant mice treated with either CD25 mAb to deplete Tregs and/or DNA vaccination. Only Treg depletion followed by neu DNA vaccination abrogated tolerance to neu, resulting in complete regression of neu(+) tumors, as well as long-term protection from spontaneous tumorigenesis in 58% of mice. The risk of developing EAT was then assessed by incorporated mTg immunization with or without LPS as adjuvant. In mice with induced tumor regression, mTg response was enhanced with modest increases in EAT development. Therefore, tumor regression induced by Treg depletion and DNA vaccination can exacerbate autoimmunity, which warrants close monitoring during immunotherapy.


Assuntos
Vacinas Anticâncer/administração & dosagem , Depleção Linfocítica , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/prevenção & controle , Receptor ErbB-2/genética , Linfócitos T Reguladores/imunologia , Tireoidite Autoimune/genética , Vacinas de DNA/administração & dosagem , Animais , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Feminino , Predisposição Genética para Doença , Depleção Linfocítica/métodos , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Células NIH 3T3 , Ratos , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/imunologia , Indução de Remissão , Linfócitos T Reguladores/patologia , Tireoglobulina/administração & dosagem , Tireoglobulina/genética , Tireoglobulina/imunologia , Tireoidite Autoimune/imunologia , Tireoidite Autoimune/prevenção & controle , Vacinas de DNA/imunologia
7.
Clin Cancer Res ; 26(11): 2515-2523, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32071118

RESUMO

PURPOSE: Preclinical data provide evidence for synergism between HER2-targeted peptide vaccines and trastuzumab. The efficacy of this combination was evaluated in patients with HER2 low-expressing breast cancer in the adjuvant setting. PATIENTS AND METHODS: A phase IIb, multicenter, randomized, single-blinded, controlled trial enrolled disease-free patients after standard therapy completion (NCT01570036). Eligible patients were HLA-A2, A3, A24, and/or A26+, and had HER2 IHC 1+/2+, FISH nonamplified breast cancer, that was node positive and/or hormone receptor-negative [triple-negative breast cancer (TNBC)]. Patients received trastuzumab for 1 year and were randomized to placebo (GM-CSF, control) or nelipepimut-S (NPS) with GM-CSF. Primary outcome was 24-month disease-free survival (DFS). Secondary outcomes were 36-month DFS, safety, and immunologic response. RESULTS: Overall, 275 patients were randomized; 136 received NPS with GM-CSF, and 139 received placebo with GM-CSF. There were no clinicopathologic differences between groups. Concurrent trastuzumab and NPS with GM-CSF was safe with no additional overall or cardiac toxicity compared with control. At median follow-up of 25.7 (interquartile range, 18.4-32.7) months, estimated DFS did not significantly differ between NPS and control [HR, 0.62; 95% confidence interval (CI), 0.31-1.25; P = 0.18]. In a planned exploratory analysis of patients with TNBC, DFS was improved for NPS versus control (HR, 0.26; 95% CI, 0.08-0.81, P = 0.01). CONCLUSIONS: The combination of NPS with trastuzumab is safe. In HER2 low-expressing breast cancer, no significant difference in DFS was seen in the intention-to-treat analysis; however, significant clinical benefit was seen in patients with TNBC. These findings warrant further investigation in a phase III randomized trial.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Adulto , Neoplasias da Mama/patologia , Feminino , Seguimentos , Humanos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/patologia , Fragmentos de Peptídeos/administração & dosagem , Prognóstico , Receptor ErbB-2/administração & dosagem , Método Simples-Cego , Taxa de Sobrevida , Trastuzumab/administração & dosagem
8.
BMJ Open Qual ; 9(4)2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33023906

RESUMO

The demand for systemic anticancer treatment continues to rise due to the increasing numbers being diagnosed with cancer and developments in treatment options. The net result is greater pressure on services and subsequent patient delays. Delays in treatment could decrease the benefit of the therapy and be detrimental to patient experience. Patients with human epidermal growth factor receptor-2 (HER 2) positive breast cancer within Raigmore Hospital waited an average of 41 min from the scheduled appointment time despite the administration of subcutaneous (SC) trastuzumab being scheduled for a 15 min treatment window. Given the frequency of these injections, this was having an adverse impact on patients and services. The aim of this project was for patients with breast cancer to receive treatment within the 15 min window. Lean principles were applied to reduce waste and increase value. Exploration of the problem led to the solution of relocating the administration of SC trastuzumab from the Macmillan Day Bed Unit (MDBU) to the Highland Breast Centre (HBC). Multiple improvement tools and techniques were used to implement the change. Data were collected on the median number of treatment episodes of SC trastuzumab per week at baseline and patient wait from appointment time to treatment completion was calculated at baseline and as an ongoing measure. Patient experience feedback was gathered following relocation of the treatment. Following relocation, the average time from scheduled appointment to discharge was 14 min (66% reduction). Patient experience feedback was positive and identified an unanticipated outcome; the regular Friday afternoon clinic, identified as most efficient for the service, was found by patients to be particularly convenient for their own planning. Through the application of Lean principles, the service was redesigned in a cost neutral way and resulted in a reduction in the wait time for treatment.


Assuntos
Antineoplásicos/administração & dosagem , Tempo para o Tratamento/normas , Listas de Espera , Instituições de Assistência Ambulatorial/organização & administração , Instituições de Assistência Ambulatorial/normas , Instituições de Assistência Ambulatorial/estatística & dados numéricos , Antineoplásicos/uso terapêutico , Agendamento de Consultas , Humanos , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/uso terapêutico , Fatores de Tempo , Tempo para o Tratamento/estatística & dados numéricos , Trastuzumab/administração & dosagem , Trastuzumab/uso terapêutico
9.
Breast Cancer Res Treat ; 115(1): 29-41, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18481173

RESUMO

The clinical use of Pseudomonas exotoxin A (PE)-based immunotoxins is limited by the toxicity and immunogenicity of PE. To overcome the limitations, we have developed PE38KDEL-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles conjugated with Fab' fragments of a humanized anti-HER2 monoclonal antibody (rhuMAbHER2). The PE38KDEL-loaded nanoparticles-anti-HER2 Fab' bioconjugates (PE-NP-HER) were constructed modularly with Fab' fragments of rhuMAbHER2 covalently linked to PLGA nanoparticles containing PE38KDEL. Compared with nontargeted nanoparticles that lack anti-HER2 Fab', PE-NP-HER specifically bound to and were sequentially internalized into HER2 overexpressing breast cancer cells, which result in significant cytotoxicity in vitro. In HER2 overexpressing tumor xenograft model system, administration of PE-NP-HER showed a superior efficacy in inhibiting tumor growth compared with PE-HER referring to PE38KDEL conjugated directly to rhuMAbHER2. Moreover, PE-NP-HER was well tolerated in mice with a higher LD(50) (LD(50) of 6.86 +/- 0.47 mg/kg vs. 2.21 +/- 0.32 mg/kg for PE-NP-HER vs. PE-HER (mean +/- SD); n = 3), and had no influence on the plasma level of plasma alanine aminotransferase (ALT) of animals when injected at a dose of 1 mg/kg where PE-HER caused significant increase of serum ALT in the treated mice. Notably, PE-NP-HER was of low immunogenicity in development of anti-PE38KDEL neutralizing antibodies and was less susceptible to inactivation by anti-PE38KDEL antibodies compared with PE-HER. This novel bioconjugate, PE-NP-HER, may represent a useful strategy for cancer treatment.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Imunotoxinas/química , Nanopartículas/química , Nanopartículas/uso terapêutico , Receptor ErbB-2/uso terapêutico , Alanina Transaminase/sangue , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Progressão da Doença , Relação Dose-Resposta a Droga , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/administração & dosagem , Nanotecnologia/métodos , Transplante de Neoplasias , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/imunologia
10.
J Drug Target ; 26(9): 840-844, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29521129

RESUMO

The use of peptides in drug development has been hampered by their poor pharmaceutical properties, most notably their inability to reliably permeate biological membranes and lack of targeting. To overcome these disadvantages, the AMino acid Intracellular Delivery SysTem (AMIDST) was developed. This modular peptide-based delivery system confers cellular permeability and organelle-specific targeting for therapeutic peptides. As demonstrated in this study, the delivery of a HER2-binding peptide to the secretory organelles of breast cancer cells resulted in intracellular sequestration, a reduction in downstream signalling, and reduced viability compared to the delivery of a control peptide. Given its modular design and ease of production, AMIDST has the potential to enhance the use of peptides as therapeutic agents.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Peptídeos/administração & dosagem , Receptor ErbB-2/administração & dosagem , Frações Subcelulares/metabolismo , Humanos , Peptídeos/química , Receptor ErbB-2/química
11.
J Drug Target ; 26(4): 365-372, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28972792

RESUMO

E75 (HER-2/neu-369-377), is an immunogenic peptide which is highly expressed in breast cancer patients. The purpose of this study was to develop an effective vaccine delivery/adjuvant system by attachment of this peptide to the surface of liposomes consisting of phospholipids including distearoylphosphocholine (DSPC) and distearoyl phosphoglycerol (DSPG) with high transition temperature (Tm) and dioleoylphosphatidylethanolamine (DOPE) (a pH-sensitive lipid for cytosolic antigen delivery) to improve antitumour immune activity against the E75 peptide. For this purpose, the E75 peptide was incorporated into liposomes consisting of DSPC/DSPG/cholesterol (Chol)/DOPE (15/2/3/5 molar ratio) through conjugation with distearoylphosphoethanolamine-N-[maleimide(polyethylene glycol)-2000] (maleimide-PEG2000-DSPE). Immunization of BALB/c mice was performed three times with different forms of liposomal formulations at 2-week intervals and antitumour immunity responses were evaluated. Results of ELISpot and flow cytometry analysis showed that mice vaccinated with DSPC/DSPG/Chol/DOPE/E75 have significantly enhanced the antigen-specific IFN-γ response of CD8+ T cells and generated cytotoxic T lymphocytes (CTL) antitumour responses. CTL responses induced by this formulation resulted in inhibition of tumour progression and longer survival time in the mice TUBO tumour model. The results revealed that the liposomes consist of DSPC/DSPG/Chol/DOPE could be suitable candidates for vaccine delivery of E75 peptide for the prevention and therapy of HER2-positive breast cancer and merit further investigation.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Vacinas Anticâncer/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Fosfolipídeos/química , Receptor ErbB-2/administração & dosagem , Animais , Neoplasias da Mama/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Progressão da Doença , Feminino , Interferon gama/imunologia , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas , Fragmentos de Peptídeos/imunologia , Receptor ErbB-2/imunologia , Taxa de Sobrevida , Linfócitos T Citotóxicos/imunologia , Temperatura de Transição
12.
Eur J Pharmacol ; 831: 87-93, 2018 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-29753042

RESUMO

The E75 peptide vaccine, derived from tumor-associated antigen HER2, is the most frequently studied anti-HER2 vaccination strategy for the treatment of breast cancer patients. It has been investigated in the several phases Ι/Π of the clinical trials and is currently being evaluated in a randomized multicenter phase III clinical trial. We conducted a systematic review and meta-analysis to clarify the outcomes of the E75 peptide vaccine including the therapeutic efficacy, the disease recurrence, the survival rate, and the side effects. Three peer-reviewed literature databases including the PubMed, Web of Science, and Scopus were sought. Of 29 trials assessed for eligibility, 16 were considered based on our inclusion criteria. Statistical analyses were performed by The Excel and STATA v.11.0. Meta-analysis of delayed-type hypersensitivity)DTH( reactions and CD8+-T cell levels, as immune responses, displayed the significant differences in the vaccinated groups compared to their non-vaccinated counterparts. In addition, the recurrence, and the overall and the disease-free survival were significantly different in the vaccinated subjects versus the control. Evaluation of the local and systemic toxicity of the E75 peptide vaccine demonstrated the minimal side effects. It seems that the E75 peptide vaccine is safe and effective, and can be used for further randomized clinical trials.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Vacinas Anticâncer/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Receptor ErbB-2/administração & dosagem , Neoplasias da Mama/imunologia , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/imunologia , Progressão da Doença , Intervalo Livre de Doença , Feminino , Humanos , Recidiva Local de Neoplasia , Fragmentos de Peptídeos/efeitos adversos , Fragmentos de Peptídeos/imunologia , Receptor ErbB-2/imunologia , Fatores de Tempo , Resultado do Tratamento
13.
Clin Cancer Res ; 24(21): 5292-5304, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30037817

RESUMO

Purpose: Response to a complex trastuzumab-based regimen is affected by multiple features of the tumor and its microenvironment. Developing a predictive algorithm is key to optimizing HER2-targeting therapy.Experimental Design: We analyzed 137 pretreatment tumors with mRNA-seq and DNA exome sequencing from CALGB 40601, a neoadjuvant phase III trial of paclitaxel plus trastuzumab with or without lapatinib in stage II to III HER2-positive breast cancer. We adopted an Elastic Net regularized regression approach that controls for covarying features within high-dimensional data. First, we applied 517 known gene expression signatures to develop an Elastic Net model to predict pCR, which we validated on 143 samples from four independent trials. Next, we performed integrative analyses incorporating clinicopathologic information with somatic mutation status, DNA copy number alterations (CNA), and gene signatures.Results: The Elastic Net model using only gene signatures predicted pCR in the validation sets (AUC = 0.76). Integrative analyses showed that models containing gene signatures, clinical features, and DNA information were better pCR predictors than models containing a single data type. Frequently selected variables from the multiplatform models included amplifications of chromosome 6p, TP53 mutation, HER2-enriched subtype, and immune signatures. Variables predicting resistance included Luminal/ER+ features.Conclusions: Models using RNA only, as well as integrated RNA and DNA models, can predict pCR with improved accuracy over clinical variables. Somatic DNA alterations (mutation, CNAs), tumor molecular subtype (HER2E, Luminal), and the microenvironment (immune cells) were independent predictors of response to trastuzumab and paclitaxel-based regimens. This highlights the complexity of predicting response in HER2-positive breast cancer. Clin Cancer Res; 24(21); 5292-304. ©2018 AACR.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Receptor ErbB-2/genética , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Biomarcadores Tumorais , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/mortalidade , Biologia Computacional/métodos , Variações do Número de Cópias de DNA , Resistencia a Medicamentos Antineoplásicos , Feminino , Perfilação da Expressão Gênica , Genômica/métodos , Humanos , Mutação , Terapia Neoadjuvante , Prognóstico , Receptor ErbB-2/administração & dosagem , Transcriptoma , Trastuzumab/administração & dosagem , Resultado do Tratamento
14.
Cancer Lett ; 430: 79-87, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-29778570

RESUMO

Human epidermal growth factor receptor 2 (HER2) is an attractive target for cancer vaccine. However, autoimmune tolerance prevents vaccines based on HER2 protein from inducing long-lasting, highly effective anti-tumor immunity. In this study, we proved that the introduction of p-nitrophenylalanine in the universal T cell epitope (named NitraTh) enhances humoral immunity induced by B cell epitope and cellular immunity induced by CTL epitope. Moreover, this NitraTh epitope can work in both mouse and human immune system. When fused with extracellular domain 23-83 of HER2, NitraTh epitope help to break the self-tolerance of HER2 and induced strong HER2 specific humoral immunity and cellular immunity. Vaccination with HER2-NitraTh can significantly inhibit the growth of HER2+B16F10 tumor cells. These findings have important implications for developing therapeutic cancer vaccines.


Assuntos
Neoplasias da Mama/terapia , Vacinas Anticâncer/imunologia , Epitopos de Linfócito T/imunologia , Imunogenicidade da Vacina , Receptor ErbB-2/imunologia , Animais , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Epitopos de Linfócito T/química , Epitopos de Linfócito T/genética , Feminino , Humanos , Tolerância Imunológica/efeitos dos fármacos , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/imunologia , Imunidade Humoral/efeitos dos fármacos , Imunidade Humoral/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Nitratos/química , Domínios Proteicos/genética , Domínios Proteicos/imunologia , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Resultado do Tratamento , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
15.
Biochim Biophys Acta ; 1725(2): 182-9, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16126344

RESUMO

The elimination of cancer cells requires strong cellular immune responses, and these responses are induced by the activation of Th1 lymphocytes. In this work, the possibility of inducing a Th1 type of immune response in vivo by mixing a HER-2/neu synthetic CTL (cytotoxic T lymphocyte) peptide [HER-2/neu (789-797)], with poly-lactide (PLA) microspheres was investigated. Various formulations of the peptide were administered to HLA-A2.1 transgenic (HHD) mice. Cellular experiments, assessing proliferation and cytokine determination in splenocyte culture supernatants, were carried out in order to evaluate the type of immune response to the antigen. The in vivo administration of the peptide antigen admixed with the PLA microspheres induced a potent immune response which was comparable to that induced by the combination of the antigen in complete Freund's adjuvant (CFA). Furthermore, the cytokine profile produced by the T lymphocytes of the immunized animals indicated that the combination of the peptide antigen with the PLA microspheres induced a strong Th1 biased immune response to the antigen. The time of peptide incubation with the microspheres prior to administration did not affect the immune response, which further simplifies the preparation of this type of vaccine. The results justify further investigation of the possibility of inducing effective cellular immune responses against cancer cells overexpressing HER-2/neu molecules by simply mixing appropriate HER-2/neu peptide antigens with PLA microspheres.


Assuntos
Portadores de Fármacos/química , Poliésteres/química , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/imunologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Vacinas/imunologia , Animais , Materiais Revestidos Biocompatíveis/administração & dosagem , Materiais Revestidos Biocompatíveis/química , Difusão , Portadores de Fármacos/administração & dosagem , Epitopos/imunologia , Imunização/métodos , Teste de Materiais , Camundongos , Microesferas , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Ligação Proteica , Receptor ErbB-2/química , Vacinas/uso terapêutico
16.
Int J Oncol ; 48(4): 1369-78, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26892612

RESUMO

Vaccination with priming and expansion of tumour reacting T cells is an important therapeutic option to be used in combination with novel checkpoint inhibitors to increase the specificity of the T cell infiltrate and the efficacy of the treatment. In this phase I/II study, 14 high-risk disease-free ovarian (OC) and breast cancer (BC) patients after completion of standard therapies were vaccinated with MUC1, ErbB2 and carcinoembryonic antigen (CEA) HLA-A2+-restricted peptides and Montanide. Patients were subjected to 6 doses of vaccine every two weeks and a recall dose after 3 months. ECOG grade 2 toxicity was observed at the injection site. Eight out of 14 patients showed specific CD8+ T cells to at least one antigen. None of 4 patients vaccinated for compassionate use showed a CD8 activation. An OC patient who suffered from a lymph nodal recurrence, showed specific anti-ErbB2 CD8+ T cells in the bulky aortic lymph nodes suggesting homing of the activated T cells. Results confirm that peptide vaccination strategy is feasible, safe and well tolerated. In particular OC patients appear to show a higher response rate compared to BC patients. Vaccination generates a long-lasting immune response, which is strongly enhanced by recall administrations. The clinical outcome of patients enrolled in the trial appears favourable, having registered no deceased patients with a minimum follow-up of 8 years. These promising data, in line with the results of similar studies, the high compliance of patients observed and the favourable toxicity profile, support future trials of peptide vaccination in clinically disease-free patients who have completed standard treatments.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Antígeno Carcinoembrionário/administração & dosagem , Mucina-1/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Receptor ErbB-2/administração & dosagem , Adulto , Idoso , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Antígeno Carcinoembrionário/imunologia , Intervalo Livre de Doença , Feminino , Citometria de Fluxo , Antígeno HLA-A2/genética , Antígeno HLA-A2/imunologia , Humanos , Imunoterapia/métodos , Linfonodos/imunologia , Linfonodos/patologia , Pessoa de Meia-Idade , Mucina-1/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/imunologia , Receptor ErbB-2/imunologia , Linfócitos T/imunologia
17.
Appl Biochem Biotechnol ; 176(4): 1029-43, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25906688

RESUMO

Human epidermal growth factor receptor 2 (HER2) is a member of the epidermal growth factor receptor (EGFR) family. In this study, the whole extracellular domain gene of HER2 was amplified by RT-PCR from human breast cancer cell line SK-BR-3. The genes of membrane-distal region (A) and membrane proximal region (B) of HER2 extracellular domain were amplified from the cloned template, and then inserted into the expression vector pET-28a and pET-30a, respectively. The recombinant expression vectors were transformed into Escherichia coli BL21 (DE3) cells and induced by isopropyl-b-D-thiogalactopyranoside (IPTG) for expression of proteins His-A and His-B. The expressed proteins were detected by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and western blot. The optimization of culture conditions led us to accomplish the recombinant protein induction with 1.0 mM IPTG at 37 °C for 8 h, and both proteins were expressed in the insoluble form. Both proteins were purified under the denaturing condition using Ni-NTA sepharose column. Balb/c mice were immunized with the purified proteins and then effectively produced polyclonal antibodies, which reached to a relatively high titer by ELISA testing and had good specificity by western blot detection. The HER2 ECD proteins His-A and His-B could be expressed in E. coli and were suitable for production of high titer antibodies against HER2 ECD.


Assuntos
Anticorpos/isolamento & purificação , Escherichia coli/genética , Vetores Genéticos/química , Glândulas Mamárias Humanas/metabolismo , Receptor ErbB-2/genética , Animais , Western Blotting , Linhagem Celular Tumoral , Clonagem Molecular , Eletroforese em Gel de Poliacrilamida , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Feminino , Expressão Gênica , Vetores Genéticos/metabolismo , Humanos , Imunização , Isopropiltiogalactosídeo/farmacologia , Glândulas Mamárias Humanas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Terciária de Proteína , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/biossíntese , Receptor ErbB-2/isolamento & purificação , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Transformação Bacteriana
18.
J Interferon Cytokine Res ; 22(5): 583-92, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12060497

RESUMO

HER-2/neu peptides have recently been shown to induce a proliferative response by peripheral CD4(+) T cells in breast cancer patients. To investigate potential differences in the local cellular immune response between breast cancer patients with and without nodal metastases, lymphocytes were isolated from axillary lymph nodes from patients with breast cancer, and proliferative and cytokine responses to HER-2/neu peptides were determined. Freshly isolated lymphocytes from lymph nodes of 7 women undergoing surgery for invasive breast cancer were plated at 20 x 10(5) cells per well in triplicate. Cells were stimulated with HER-2/neu peptides at 50 microg/ml and with control antigens. Incorporation of tritium-labeled thymidine was determined 4 days later. The levels of the cytokines interferon-gamma (IFN-gamma), interleukin-4 (IL-4), and IL-10 were determined at priming and at restimulation with HER-2/neu peptides using a cytokine-specific, double-sandwich, enzyme-linked immunosorbent assay (ELISA). Lymphocytes isolated from the axillary lymph nodes of the patients mounted significant cellular immune response to HER-2/neu peptides, manifested by proliferation and specific cytokine elaboration. Proliferative responses to HER-2/neu peptides were seen in lymphocytes of patients with and without overexpression of HER-2/neu in the primary tumor. In some patients, the proliferative response to HER-2/neu peptides in lymphocytes from lymph nodes with metastases was absent or blunted compared with the response in lymphocytes from lymph nodes without metastases from the same patient (p < 0.05). HER-2/neu peptides induced a predominantly T helper type 1 (Th1) pattern of cytokine response in nodal lymphocytes isolated from breast cancer patients. A Th1-specific cytokine production pattern was maintained at priming and restimulation with HER-2/neu peptides and was amplified with IL-12 costimulation. These results indicate that HER-2/neu peptides can activate T cells in draining lymph nodes from women with invasive breast cancer. This activation is associated with a predominantly Th1 cytokine response, which suggests that conditioning with HER-2/neu peptides may be of value in the development of breast cancer vaccines.


Assuntos
Neoplasias da Mama/imunologia , Linfonodos/imunologia , Receptor ErbB-2/imunologia , Antígenos de Neoplasias , Axila , Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/imunologia , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/secundário , Citocinas/biossíntese , Feminino , Humanos , Imunidade Celular , Técnicas In Vitro , Interleucina-12/administração & dosagem , Linfonodos/metabolismo , Metástase Linfática/imunologia , Ativação Linfocitária , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/metabolismo , Células Th1/imunologia
19.
Cancer Immun ; 3: 16, 2003 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-14629132

RESUMO

In this study, we identified a c-erbB-2/HER2/neu (HER2)-derived Th epitope (HER2 (16-30) ) and examined the role of Th epitopes in HER2-specific CD8+ T cell induction and in vivo tumor eradication, with a particular emphasis on the role of tumor cell-derived Th epitopes. Immunization of BALB/c mice using a mixture of Th epitope HER2 (16-30) and CTL epitope HER2 (63-71) administered subcutaneously with murine GM-CSF (mGM-CSF) induced a much higher level of HER2 (63-71) -specific CD8+ T cells compared with that obtained with the CTL epitope alone. HER2-unrelated OVA-derived Th epitope (OVA (323-339) ) exhibited a similar enhancing effect on HER2 (63-71) -specific CD8+ T cell induction. However, only mice immunized with HER2 (16-30) and HER2 (63-71), but not with a tumor-unrelated OVA (323-339) and HER2 (63-71), showed in vivo eradication of CMS5mHE tumor cells expressing HER2 but not OVA. This distinction was observed in preventative as well as therapeutic experimental settings. Conversely, both HER2 (16-30) and OVA (323-339) Th epitopes were equally effective in inducing the eradication of CMS5mHEOVA tumor cells which express HER2 as well as OVA. Our results clearly indicate that CTL and Th epitopes of target tumor cell origin should be used for effective induction of in vivo antitumor immunity.


Assuntos
Linfócitos T CD8-Positivos/efeitos dos fármacos , Vacinas Anticâncer/administração & dosagem , Ativação Linfocitária/efeitos dos fármacos , Neoplasias Experimentais/prevenção & controle , Receptor ErbB-2/administração & dosagem , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Peptídeos/administração & dosagem , Peptídeos/imunologia , Receptor ErbB-2/imunologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Cancer Chemother Pharmacol ; 46 Suppl: S77-82, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10950153

RESUMO

Genetic changes leading to protooncogene activation qualitatively and/or quantitatively alter their gene products and are exclusively or largely restricted to transforming cells and their precursors. The overexpression of HER2 is among those changes and is often detected in adenocarcinomas such as breast, ovarian, lung, and gastric cancer. This provides a rationale for exploring the possibility that HER2 is a target of host immune responses against cancer cells. We have recently demonstrated that HER2 can be a target for tumor-rejecting immune responses against syngeneic murine HER2+ tumor cells. We defined two different peptides, HER2p63-71 and HER2p780-788, with a Kd anchor motif that can induce CD8+ cytotoxic T lymphocytes (CTLs). The growth of HER2+ syngeneic tumors was suppressed in mice immunized with HER2p63-71 or p780-788. Since murine Kd and human HLA-A24 share a similar anchor motif for peptides, HER2p63 71 and HER2p780-788 were examined for induction of CTLs in HLA-A24+ individuals. CD8+ CTL clones specific for these peptides were established and they lysed HER2+ tumor cells in a human leukocyte antigen (HLA)-A24-restricted manner. To elicit specific CD8+ T cell immune responses against cancer, the development of efficient devices to deliver tumor antigen peptides to the major histocompatibility complex (MHC) class I pathway constitutes a central issue. We have developed a novel formula of hydrophobized polysaccharide nanoparticles which can deliver a HER2 oncoprotein containing an epitope peptide to the MHC class I pathway. We designed a simple protein delivery system: cholesteryl group-bearing polysaccharides, mannan or pullulan (CHM or CHP, respectively), complexed with the truncated HER2 protein containing the 147 N-terminal amino acids. These complexes were able to induce CD8+ CTLs against HER2+ tumors. CTLs were MHC class I restricted and specifically recognized HER2p63-71, a part of a truncated HER2 protein used as an immunogen. The complete rejection of tumors also occurred when CHM-HER2 was applied early after tumor implantation. In the effector phase of in vivo tumor rejection, CD8+ T cells played a major role. The results suggest that this unique hydrophobized polysaccharide may help soluble proteins to induce cellular immunity. Such a novel vaccine may be of potential benefit in cancer prevention and cancer therapy.


Assuntos
Vacinas Anticâncer/imunologia , DNA Complementar/genética , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Células Dendríticas/imunologia , Feminino , Fibrossarcoma/genética , Fibrossarcoma/imunologia , Glucanos/administração & dosagem , Humanos , Ativação Linfocitária/imunologia , Mananas/administração & dosagem , Sarcoma de Mastócitos/genética , Sarcoma de Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Receptor ErbB-2/administração & dosagem , Linfócitos T Reguladores/imunologia , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA