Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Br J Cancer ; 125(2): 152-154, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33772155

RESUMEN

We developed a PD-1 B-cell epitope vaccine (PD1-Vaxx) to rival nivolumab therapy which has received ethics approvals for a Phase 1 clinical trial in Australia. The US FDA granted Investigational New Drug approval to Imugene Ltd for clinical testing in NSCLC. We demonstrated synergistic vaccine combinations with an HER-2 targeted vaccine (B-Vaxx).


Asunto(s)
Vacunas contra el Cáncer , Neoplasias Pulmonares , Australia , Epítopos de Linfocito B , Humanos , Nivolumab
2.
Future Oncol ; 16(23): 1767-1791, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32564612

RESUMEN

In light of the numerous US FDA-approved humanized monoclonal antibodies (mAbs) for cancer immunotherapy, it is surprising that the advancement of B-cell epitope vaccines designed to elicit a natural humoral polyclonal antibody response has not gained traction in the immune-oncology landscape. Passive immunotherapy with humanized mAbs (Trastuzumab [Herceptin®]; Pertuzumab [Perjeta®]) has provided clinical benefit to breast cancer patients, albeit with significant shortcomings including toxicity problems and resistance, high costs, sophisticated therapeutic regimen and long half-life. The role of B-cell humoral immunity in cancer is under appreciated and underdeveloped. We have advanced the idea of active immunotherapy with chimeric B-cell epitope peptides incorporating a 'promiscuous' T-cell epitope that elicits a polyclonal antibody response, which provides safe, cost-effective therapeutic advantage over mAbs. We have created a portfolio of validated B-cell peptide epitopes against multiple receptor tyrosine kinases (HER-1, HER-3, IGF-1R and VEGF). We have successfully translated two HER-2 combination B-cell peptide vaccines in Phase I and II clinical trials. We have recently developed an effective novel PD-1 vaccine. In this article, I will review our approaches and strategies that focus on B-cell epitope cancer vaccines.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/inmunología , Vacunas contra el Cáncer/uso terapéutico , Epítopos de Linfocito B/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/tratamiento farmacológico , Vacunas de Subunidad/uso terapéutico , Humanos , Neoplasias/inmunología , Pronóstico
3.
Mol Pharmacol ; 87(2): 150-61, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25391374

RESUMEN

Resistance to the human epidermal growth factor receptor (HER2)-targeted antibody trastuzumab is a major clinical concern in the treatment of HER2-positive metastatic breast cancer. Increased expression or signaling from the insulin-like growth factor-1 receptor (IGF-1R) has been reported to be associated with trastuzumab resistance. However, the specific molecular and biologic mechanisms through which IGF-1R promotes resistance or disease progression remain poorly defined. In this study, we found that the major biologic effect promoted by IGF-1R was invasion, which was mediated by both Src-focal adhesion kinase (FAK) signaling and Forkhead box protein M1 (FoxM1). Cotargeting IGF-1R and HER2 using either IGF-1R antibodies or IGF-1R short hairpin RNA in combination with trastuzumab resulted in significant but modest growth inhibition. Reduced invasion was the most significant biologic effect achieved by cotargeting IGF-1R and HER2 in trastuzumab-resistant cells. Constitutively active Src blocked the anti-invasive effect of IGF-1R/HER2 cotargeted therapy. Furthermore, knockdown of FoxM1 blocked IGF-1-mediated invasion, and dual targeting of IGF-1R and HER2 reduced expression of FoxM1. Re-expression of FoxM1 restored the invasive potential of IGF-1R knockdown cells treated with trastuzumab. Overall, our results strongly indicate that therapeutic combinations that cotarget IGF-1R and HER2 may reduce the invasive potential of cancer cells that are resistant to trastuzumab through mechanisms that depend in part on Src and FoxM1.


Asunto(s)
Neoplasias de la Mama/metabolismo , Quinasa 1 de Adhesión Focal/biosíntesis , Factores de Transcripción Forkhead/biosíntesis , Regulación Neoplásica de la Expresión Génica , Receptor ErbB-2/biosíntesis , Receptor IGF Tipo 1/biosíntesis , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Proteína Forkhead Box M1 , Genes src/fisiología , Humanos , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/genética , Transducción de Señal/fisiología
4.
J Immunol ; 191(1): 217-27, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23698748

RESUMEN

Epidermal growth factor receptor (EGFR) is a validated target for several cancers including lung, colorectal, and certain subtypes of breast cancer. Cetuximab targets ligand binding of EGFR, but major problems like high cost, short t1/2, toxicity, and emergence of resistance are associated with the drug. Immunization with EGFR B cell epitopes will train the immune system to produce specific Abs that can kill cancer cells. Also, therapy with stable, less-expensive, and nontoxic EGFR peptide mimics will block EGFR signaling and inhibit cancer growth. We designed three peptides based on the contact sites between EGF and EGFR. The B cell epitopes were synthesized alone and also linked with the measles virus T cell epitope to produce a chimeric peptide vaccine. The peptide vaccines were immunogenic in both mice and rabbits and Abs raised against the vaccine specifically bound EGFR-expressing cells and recombinant human EGFR protein. The peptide mimics and the anti-peptide Abs were able to inhibit EGFR signaling pathways. Immunization with the peptide vaccine or treatment with the B cell epitopes significantly reduced tumor growth in both transplantable breast and lung cancer models. Immunohistochemical analysis also showed significant reductions in microvascular density and actively dividing cells in the tumor sections after treatment in the FVB/n breast cancer model. The 418-435 B cell epitope was the best candidate both as a vaccine or peptide mimic because it caused significant inhibition in the two mouse models. Our results show that this novel EGFR B cell epitope has great potential to be used as a vaccine or treatment option for EGFR-expressing cancers.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Receptores ErbB/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/prevención & control , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/prevención & control , Peptidomiméticos , Animales , Vacunas contra el Cáncer/metabolismo , Línea Celular Tumoral , Epítopos de Linfocito B/administración & dosificación , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito B/metabolismo , Receptores ErbB/administración & dosificación , Receptores ErbB/antagonistas & inhibidores , Humanos , Ligandos , Neoplasias Pulmonares/inmunología , Neoplasias Mamarias Experimentales/inmunología , Ratones , Fosforilación/inmunología , Unión Proteica/inmunología , Estructura Terciaria de Proteína , Conejos , Transducción de Señal/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/metabolismo
5.
J Vis Exp ; (197)2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37486131

RESUMEN

The inhibition of checkpoint receptors (PD-1, PD-L1, and CTLA-4) with monoclonal antibodies has shown great benefit in clinical trials for treating cancer patients and has become a mainstay approach in modern cancer immunotherapy. However, only a subset of patients respond to checkpoint monoclonal antibody immunotherapy. Therefore, it is urgent to develop new therapeutic strategies against cancer. A novel B-cell peptide epitope PDL1 (programmed death ligand 1) cancer vaccine has been developed, with amino acids 130-147 linked to the MVF peptide ("promiscuous" T-cell measles virus fusion protein) via a GPSL linker. Preclinical testing has indicated that this PDL1 vaccine (PDL1-Vaxx) effectively stimulates highly immunogenic antibodies in animals. Animals immunized with PDL1-Vaxx show reduced tumor burden and extended survival rates in various animal cancer models. The mechanisms of action indicate that vaccine-elicited antibodies inhibit tumor cell proliferation, induce apoptosis, and block the PD-1/PD-L1 interaction. This manuscript introduces a magnetic bead-based assay that uses a dual-reporter flow analysis system to evaluate the PD-1/PD-L1 interaction and its blockade by the anti-PDL1 antibodies raised against the PDL1-Vaxx.


Asunto(s)
Neoplasias , Vacuna contra Viruela , Animales , Antígeno B7-H1 , Receptor de Muerte Celular Programada 1 , Vacuna contra Viruela/uso terapéutico , Neoplasias/tratamiento farmacológico , Anticuerpos Monoclonales , Péptidos , Vacunas de Subunidad/uso terapéutico , Fenómenos Magnéticos , Inmunoterapia
6.
JAMA Otolaryngol Head Neck Surg ; 149(2): 168-176, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36580281

RESUMEN

Importance: Squamous cell carcinoma of the head and neck (HNSCC) is prevalent globally and in the US. Management, particularly after disease recurrence, can be challenging, and exploring additional treatment modalities, such as therapeutic cancer vaccines, may offer an opportunity to improve outcomes in this setting. Observations: This review provides an overview of the clinical efficacy of different treatment modalities that are currently available for the treatment of recurrent and metastatic HNSCC, including checkpoint inhibitors and targeted therapies, with a detailed summary of the numerous T-cell vaccines that have been studied in the setting of HNSCC, as well as a detailed summary of B-cell therapeutic vaccines being investigated for various malignant tumors. Conclusions and Relevance: The findings of this review suggest that several therapeutic T-cell and B-cell vaccines, which have been recently developed and evaluated in a clinical setting, offer a promising treatment modality with the potential to improve outcomes for patients with recurrent and metastatic HNSCC.


Asunto(s)
Vacunas contra el Cáncer , Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Vacunas contra el Cáncer/uso terapéutico , Recurrencia Local de Neoplasia/terapia , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas/patología
7.
J Biol Chem ; 286(15): 13612-25, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21321115

RESUMEN

Angiogenesis, or formation of new blood vessels, is crucial to cancer tumor growth. Tumor growth, progression, and metastasis are critically influenced by the production of the pro-angiogenic vascular endothelial growth factor (VEGF). Promising anti-angiogenic drugs are currently available; however, their susceptibilities to drug resistance and long term toxicity are serious impediments to their use, thus requiring the development of new therapeutic approaches for safe and effective angiogenic inhibitors. In this work, peptides were designed to mimic the VEGF-binding site to its receptor VEGFR-2. The VEGF conformational peptide mimic, VEGF-P3(CYC), included two artificial cysteine residues, which upon cyclization constrained the peptide in a loop native-like conformation to better mimic the anti-parallel structure of VEGF. The engineered cyclic VEGF mimic peptide demonstrated the highest affinity to VEGFR-2 by surface plasmon resonance assay. The VEGF peptide mimics were evaluated as inhibitors in several in vitro assays in which VEGF-dependent signaling pathways were observed. All VEGF mimics inhibited VEGFR-2 phosphorylation with VEGF-P3(CYC) showing the highest inhibitory effects when compared with unstructured peptides. Additionally, we show in several angiogenic in vitro assays that all the VEGF mimics inhibited endothelial cell proliferation, migration, and network formation with the conformational VEGF-P3 (CYC) being the best. The VEGF-P3(CYC) also caused a significant delay in tumor development in a transgenic model of VEGF(+/-)Neu2-5(+/-). These results indicate that the structure-based design is important for the development of this peptidomimetic and for its anti-angiogenic effects.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Materiales Biomiméticos/farmacología , Células Endoteliales/metabolismo , Péptidos/farmacología , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular , Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/química , Animales , Sitios de Unión , Materiales Biomiméticos/síntesis química , Materiales Biomiméticos/química , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Ratones , Ratones Transgénicos , Péptidos/síntesis química , Péptidos/química , Fosforilación/efectos de los fármacos , Conejos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
J Biol Chem ; 286(15): 13626-37, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21325276

RESUMEN

HER-2 is a member of the EGF receptor family and is overexpressed in 20-30% of breast cancers. HER-2 overexpression causes increased expression of VEGF at both the RNA and protein levels. HER-2 and VEGF are therefore considered good targets for cancer treatment, which has led to the development of two humanized monoclonal antibodies (mAb) pertuzumab and bevacizumab. Although passive immunotherapy with these Abs are approved for treatment of advanced breast cancer, a number of concerns exist. Treatment is expensive, has a limited duration of action, and is usually accompanied by serious side effects. We hypothesized that therapy with conformational peptide mimics aimed at blocking receptor-ligand interaction is potentially safer with little toxicity, cheaper with a longer half-life, and has greater penetrating abilities than mAbs. We designed and synthesized peptides based on the binding of HER-2 with pertuzumab and VEGF with VEGFR2. We show that treatment with the peptide mimics induces potent anti-tumor responses in vitro as determined by cell viability, proliferation, and HER2 phosphorylation assays. We also demonstrate in a transplantable BALB/c mouse tumor model that treatment with the peptide mimics resulted in a greater delay in tumor growth and development. Similarly, treatment with the peptide mimics inhibited angiogenesis in vivo as assessed by a Matrigel plug assay. To address the problem of degradability of L-amino acid peptides in vivo, we synthesized the retro-inverso D-peptide mimics that resulted in higher efficacy in treatment. Our study shows that combination treatment with HER-2 and VEGF peptide mimics provides greater efficacy than individual treatments.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Materiales Biomiméticos/farmacocinética , Neoplasias Experimentales/tratamiento farmacológico , Péptidos/farmacología , Receptor ErbB-2 , Factor A de Crecimiento Endotelial Vascular , Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/química , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/química , Materiales Biomiméticos/síntesis química , Materiales Biomiméticos/química , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Péptidos/síntesis química , Péptidos/química
9.
Future Oncol ; 8(8): 961-87, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22894670

RESUMEN

The ErbB family (HER-1, HER-2, HER-3 and HER-4) of receptor tyrosine kinases has been the focus of cancer immunotherapeutic strategies while antiangiogenic therapies have focused on VEGF and its receptors VEGFR-1 and VEGFR-2. Agents targeting receptor tyrosine kinases in oncology include therapeutic antibodies to receptor tyrosine kinase ligands or the receptors themselves, and small-molecule inhibitors. Many of the US FDA-approved therapies targeting HER-2 and VEGF exhibit unacceptable toxicities, and show problems of efficacy, development of resistance and unacceptable safety profiles that continue to hamper their clinical progress. The combination of different peptide vaccines and peptidomimetics targeting specific molecular pathways that are dysregulated in tumors may potentiate anticancer immune responses, bypass immune tolerance and circumvent resistance mechanisms. The focus of this review is to discuss efforts in our laboratory spanning two decades of rationally developing peptide vaccines and therapeutics for breast cancer. This review highlights the prospective benefit of a new, untapped category of therapies biologically targeted to EGF receptor (HER-1), HER-2 and VEGF with potential peptide 'blockbusters' that could lay the foundation of a new paradigm in cancer immunotherapy by creating clinical breakthroughs for safe and efficacious cancer cures.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Neoplasias/terapia , Vacunas de Subunidad/uso terapéutico , Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Vacunas contra el Cáncer/farmacología , Modelos Animales de Enfermedad , Epítopos de Linfocito B/inmunología , Receptores ErbB/química , Receptores ErbB/inmunología , Humanos , Inmunoterapia , Imitación Molecular/inmunología , Terapia Molecular Dirigida , Neoplasias/inmunología , Neoplasias/metabolismo , Neovascularización Patológica/terapia , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/química , Receptor ErbB-2/inmunología , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Vacunas de Subunidad/farmacología , Factores de Crecimiento Endotelial Vascular/química , Factores de Crecimiento Endotelial Vascular/inmunología
10.
Front Oncol ; 12: 826566, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35646678

RESUMEN

Immunotherapy with monoclonal antibodies to checkpoint inhibitors against the PD-1/PD-L1 signaling pathway is a landmark achievement in cancer therapy. Some anti-PD-1 inhibitors such as nivolumab and pembrolizumab have shown clinical success, in a percentage of patients with prolonged survival rates. However, adverse effects accompany these benefits. In this case, strategies with lower toxicity and increased specificity are urgently required. Cancer vaccines have the ability to stimulate the native immune system and in particular, an engineered B-cell epitope can elicit high-affinity polyclonal antibodies with similar efficacy to PD-1 monoclonal antibodies in murine animal models. We have previously designed and synthesized a unique B-cell vaccine, PD1-Vaxx [MVF-PD-1(92-110)], and we have tested the immunogenicity and antitumor properties in CT26 colon cancer BALB/c syngeneic mice model. This manuscript provides results from comprehensive preclinical pharmacology studies encompassing primary and secondary pharmacodynamics, biodistribution, and safety studies. The results from these preclinical studies support the use of PD1-Vaxx in a first-in-human clinical trial in patients with non-small cell lung cancer (NSCLC). A phase I trial in patients with NSCLC has commenced.

11.
Oncoimmunology ; 11(1): 2127691, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36211807

RESUMEN

Blockade of checkpoint receptors with monoclonal antibodies against CTLA-4, PD-1 and PD-L1 has shown great clinical success in several cancer subtypes, yielding unprecedented responses albeit a significant number of patients develop resistance and remain refractory. Both PD-1/PD-L1 and HER-2 signaling pathway inhibitors have limited efficacy and exhibits significant toxicities that limit their use. Ongoing clinical studies support the need for rationale combination of immuno-oncology agents to make a significant impact in the lives of cancer patients. We introduce the development of a novel chimeric PD-L1 B-cell peptide epitope vaccine (amino acid 130-147) linked to a "promiscuous" T cell measles virus fusion (MVF) peptide (MVF-PD-L1(130); PDL1-Vaxx) or linked to tetanus toxoid (TT3) TT3-PD-L1 (130) via a linker (GPSL). These vaccine constructs are highly immunogenic and antigenic in several syngeneic animal models. The PD-L1 vaccines elicited high titers of polyclonal antibodies that inhibit tumor growth in multiple syngeneic cancer models, eliciting antibodies of different subtypes IgG1, IgG2a, IgG2b and IgG3, induced PD-1/PD-L1 blockade, decreased proliferation, induced apoptosis and caused ADCC of tumor cells. The PDL1-Vaxx induces similar inhibition of tumor growth versus the standard anti-mouse PD-L1 antibody in both syngeneic BALB/c and C57BL/6J mouse models. The combination of PDL1-Vaxx with HER-2 vaccine B-Vaxx demonstrated synergistic tumor inhibition in D2F2/E2 carcinoma cell line. The anti-PDL1-Vaxx block PD-1/PD-L1 interaction and significantly prolonged anti-tumor responses in multiple syngeneic tumor models. The combination of HER-2 vaccine (B-Vaxx) with either PDL1-Vaxx or PD1-Vaxx demonstrated synergistic tumor inhibition. PDL1-Vaxx is a promising novel safe checkpoint inhibitor vaccine.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Aminoácidos , Animales , Anticuerpos Monoclonales , Antígeno B7-H1 , Antígeno CTLA-4 , Epítopos de Linfocito B , Inmunidad , Inmunoglobulina G , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Péptidos , Receptor de Muerte Celular Programada 1 , Toxoide Tetánico , Vacunas de Subunidad
12.
J Biol Chem ; 285(5): 3168-80, 2010 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-19940158

RESUMEN

Complex I (NQR) is a critical site of superoxide (O2-*) production and the major host of redox protein thiols in mitochondria. In response to oxidative stress, NQR-derived protein thiols at the 51- and 75-kDa subunits are known to be reversibly S-glutathionylated. Although several glutathionylated domains from NQR 51 and 75 kDa have been identified, their roles in the regulatory functions remain to be explored. To gain further insights into protein S-glutathionylation of complex I, we used two peptides of S-glutathionylated domain ((200)GAGAYICGEETALIESIEGK(219) of 51-kDa protein and (361)VDSDTLCTEEVFPTAGAGTDLR(382) of 75-kDa protein) as chimeric epitopes incorporating a "promiscuous" T-cell epitope to generate two polyclonal antibodies, AbGSCA206 and AbGSCB367. Binding of AbGSCA206 and AbGSCB367 inhibited NQR-mediated O2-* generation by 37 and 57%, as measured by EPR spin-trapping. To further provide an appropriate control, two peptides of non-glutathionylated domain ((21)SGDTTAPKKTSFGSLKDFDR(40) of 51-kDa peptide and (100)WNILTNSEKTKKAREGVMEFL(120) of 75-kDa peptide) were synthesized as chimeric epitopes to generate two polyclonal antibodies, Ab51 and Ab75. Binding of A51 did not affect NQR-mediated generation to a significant level. However, binding of Ab75 inhibited NQR-mediated O2-*generation by 35%. None of AbGSCA206, AbGSCB367, Ab51, or Ab75 showed an inhibitory effect on the electron transfer activity of NQR, suggesting that antibody binding to the glutathione-binding domain decreased electron leakage from the hydrophilic domain of NQR. When heart tissue homogenates were immunoprecipitated with Ab51 or Ab75 and probed with an antibody against glutathione, protein S-glutathionylation was enhanced in post-ischemic myocardium at the NQR 51-kDa subunit, but not at the 75-kDa subunit, indicating that the 51-kDa subunit of flavin subcomplex is more sensitive to oxidative stress resulting from myocardial infarction.


Asunto(s)
Anticuerpos/química , Complejo I de Transporte de Electrón/metabolismo , Glutatión/química , Péptidos/química , Animales , Bovinos , Espectroscopía de Resonancia por Spin del Electrón , Epítopos/química , Masculino , Mitocondrias/metabolismo , Isquemia Miocárdica/patología , Estrés Oxidativo , Estructura Terciaria de Proteína , Conejos , Ratas , Ratas Sprague-Dawley , Superóxidos
13.
Mol Med ; 17(5-6): 508-15, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21267511

RESUMEN

Deterioration of diaphragm function is one of the prominent factors that contributes to the susceptibility of serious respiratory infections and development of respiratory failure in patients with Duchenne Muscular Dystrophy (DMD). The NF-κB signaling pathway has been implicated as a contributing factor of dystrophic pathology, making it a potential therapeutic target. Previously, we demonstrated that pharmacological inhibition of NF-κB via a small NEMO Binding Domain (NBD) peptide was beneficial for reducing pathological features of mdx mice. Now, we stringently test the effectiveness and clinical potential of NBD by treating mdx mice with various formulations of NBD and use diaphragm function as our primary outcome criteria. We found that administering DMSO-soluble NBD rescued 78% of the contractile deficit between mdx and wild-type (WT) diaphragm. Interestingly, synthesis of a GLP NBD peptide as an acetate salt permitted its solubility in water, but as a negative consequence, also greatly attenuated functional efficacy. However, replacing the acetic acid counterion of the NBD peptide with trifluoroacetic acid retained the peptide's water solubility and significantly restored mdx diaphragm contractile function and improved histopathological indices of disease in both diaphragm and limb muscle. Together, these results support the feasibility of using a mass-produced, water-soluble NBD peptide for clinical use.


Asunto(s)
Diafragma/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Distrofia Muscular de Duchenne/tratamiento farmacológico , FN-kappa B/metabolismo , Péptidos/uso terapéutico , Animales , Ensayo de Cambio de Movilidad Electroforética , Femenino , Masculino , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/metabolismo , Péptidos/síntesis química , Péptidos/química
14.
J Immunol ; 182(2): 851-9, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19124728

RESUMEN

The innate immune response plays a key role as the primary host defense against invading pathogens including viruses. We have previously shown that treatment of human monocyte-derived macrophages with EBV-encoded dUTPase induces the expression of proinflammatory cytokines through the activation of NF-kappaB. However, the receptor responsible for EBV-encoded dUTPase-mediated biological effects is not known. In this study, we demonstrate that the purified EBV-encoded dUTPase activates NF-kappaB in a dose-dependent manner through TLR2 and requires the recruitment of the adaptor molecule MyD88 but not CD14. Furthermore, activation of NF-kappaB was abrogated by anti-TLR2, anti-EBV-encoded dUTPase blocking Abs and the overexpression of a dominant negative construct of MyD88 in human embryonic kidney 293 cells expressing TLR2. In addition, treatment of human monocyte-derived macrophages with the anti-EBV-encoded dUTPase Ab 7D6 or the anti-TLR2 Ab blocked the production of IL-6 by the EBV-encoded dUTPase. To our knowledge, this is the first report demonstrating that a nonstructural protein encoded by EBV is a pathogen-associated molecular pattern and that it has immunomodulatory functions. Although additional studies are necessary to define the signaling pathways activated by the EBV-encoded dUTPase and to determine its role in modulating immune responses to EBV infection, our results suggest that the dUTPase could be a potential target for the development of novel therapeutic agents against infections caused by EBV.


Asunto(s)
Herpesvirus Humano 4/genética , Herpesvirus Humano 4/inmunología , Factor 88 de Diferenciación Mieloide/fisiología , FN-kappa B/metabolismo , Pirofosfatasas/fisiología , Transducción de Señal/inmunología , Receptor Toll-Like 2/fisiología , Línea Celular , Células Cultivadas , Relación Dosis-Respuesta Inmunológica , Humanos , Receptores de Lipopolisacáridos/fisiología , Pirofosfatasas/genética , Proteínas Virales/genética , Proteínas Virales/fisiología
15.
Gynecol Oncol ; 119(3): 564-70, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20822802

RESUMEN

OBJECTIVE: To assess the role of active immunotherapy targeting VEGF with a peptide vaccine as a potential treatment for ovarian cancer. METHODS: A peptide vaccine targeting antigenic B-cell epitopes of VEGF were identified and linked to a promiscuous T-cell epitope. Elicited antibodies were assessed for their ability to recognize the VEGF protein, inhibit angiogenesis, inhibit the interaction of VEGF with its receptor, and inhibit cancer growth in mice. RESULTS: Following immunization, high-titered elicited antibodies were shown to be specific for the full-length VEGF protein by ELISA and Western blot. Anti-VEGF peptide antibodies inhibited cellular migration, proliferation, invasion, tube formation, and growth of aortic ring cultures. These antibodies inhibited the interaction between VEGF and its receptor (VEGFR2) in a concentration-dependent manner. Confirmation of this mechanism was demonstrated through inhibition of VEGFR2 phosphorylation following culture of human endothelial vein endothelial cells with anti-VEGF peptide antibodies. These antibodies were shown to inhibit ovarian cancer xenograft growth in a nude mouse model following intraperitoneal passive immunization. Active immunization with the VEGF peptide vaccine inhibited VEGF-dependent pancreatic islet cell tumor growth in RIP1-Tag2 transgenic mice and was associated with decreased vasculogenesis in these tumors compared with animals vaccinated with an irrelevant peptide. Active immunization also inhibited growth of tumors from a VEGF overexpressing ovarian cancer cell line, resulting in decreased tumor size and tumor vessel density compared with control mice. CONCLUSIONS: Active immunization with VEGF peptides elicits antibodies that inhibit tumor growth by blocking VEGF-dependent angiogenesis.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Neoplasias Ováricas/terapia , Fragmentos de Péptidos/inmunología , Vacunas de Subunidad/inmunología , Factor A de Crecimiento Endotelial Vascular/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Epítopos de Linfocito B/inmunología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Datos de Secuencia Molecular , Virus de la Neumonía Murina , Neovascularización Patológica/inmunología , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/inmunología , Fragmentos de Péptidos/síntesis química , Conejos , Vacunas de Subunidad/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/síntesis química , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Oncoimmunology ; 9(1): 1818437, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33117602

RESUMEN

Therapeutic blockade of PD-1/PD-L1 signaling with monoclonal antibodies (mAbs) has shown clinical success and activity across a broad set of cancer subtypes. However, monotherapy with PD-1/PD-L1 inhibitors are only effective in a subset of patients and ongoing studies show efficacy of treatment depends on a combinatorial approach. Contrary to mAbs chimeric B-cell cancer vaccines incorporating a "promiscuous" T-cell epitope have the advantage of producing a polyclonal B-cell antibody that can potentially induce memory B- and T-cell responses, while reducing immune evasion and suppression. Here, we describe a novel PD-1 B-cell peptide epitope vaccine (amino acid 92-110; PD1-Vaxx) linked to a measles virus fusion peptide (MVF) amino acid 288-302 via a four amino acid residue (GPSL) emulsified in Montanide ISA 720VG that aims to induce the production of polyclonal antibodies that block PD-1 signaling and thus trigger anticancer effects similar to nivolumab. In preclinical studies, the PD1-Vaxx outperformed the standard anti-mouse PD-1 antibody (mAb 29F.1A12) in a mouse model of human HER-2 expressing colon carcinoma. Furthermore, the combination of PD1-Vaxx with combo HER-2 peptide vaccine (B-Vaxx) showed enhanced inhibition of tumor growth in colon carcinoma BALB/c model challenged with CT26/HER-2 cells. The PD-1 or combined vaccines were safe with no evidence of toxicity or autoimmunity.


Asunto(s)
Vacunas contra el Cáncer , Animales , Anticuerpos Monoclonales Humanizados , Linfocitos B , Epítopos de Linfocito B , Humanos , Inmunoterapia , Ratones , Ratones Endogámicos BALB C , Receptor de Muerte Celular Programada 1 , Trastuzumab
17.
Clin Cancer Res ; 25(12): 3495-3507, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30804020

RESUMEN

PURPOSE: This first-in-human phase I study (NCT01417546) evaluated the safety profile, optimal immunologic/biological dose (OID/OBD), and immunogenicity of the combination of two peptide B-cell epitope vaccines engineered to represent the trastuzumab- and pertuzumab-binding sites. Although trastuzumab and pertuzumab have been approved for clinical use, patients often develop resistance to these therapies. We have advanced a new paradigm in immunotherapy that focuses on humoral responses based on conformational B-cell epitope vaccines. PATIENTS AND METHODS: The vaccine is comprised of two chimeric HER-2 B-cell peptide vaccines incorporating a "promiscuous T-cell epitope." Patients were immunized with the vaccine constructs emulsified with nor-muramyl-dipeptide adjuvant in a water-in-oil Montanide ISA 720VG vehicle. Eligible patients with metastatic and/or recurrent solid tumors received three inoculations every 3 weeks. RESULTS: Forty-nine patients with a median of 4 prior lines of chemotherapy received at least 1 vaccination. Twenty-eight patients completed the 3 vaccination regimens. Six patients received 1 six-month boost after the regimen, and one patient received 7 six-month boosts. No serious adverse reactions or dose-limiting toxicities were observed. The vaccine was well tolerated with dose level 2 as the recommended phase II dose. The most common related toxicity in all patients was injection-site reactions (24%). Two patients had a partial response, 14 had stable disease, and 19 had progressive disease. CONCLUSIONS: The study vaccine is safe, exhibits antitumor activity, and shows preliminary indication that peptide vaccination may avoid therapeutic resistance and offer a promising alternative to monoclonal antibody therapies.


Asunto(s)
Linfocitos B/inmunología , Epítopos de Linfocito B/inmunología , Manitol/análogos & derivados , Neoplasias/tratamiento farmacológico , Ácidos Oléicos/química , Receptor ErbB-2/inmunología , Vacunas de Subunidad/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/efectos adversos , Adyuvantes Inmunológicos/química , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos B/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Estudios de Cohortes , Femenino , Humanos , Inmunización/métodos , Masculino , Manitol/química , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/inmunología , Neoplasias/metabolismo , Receptor ErbB-2/antagonistas & inhibidores , Resultado del Tratamiento , Células Tumorales Cultivadas , Vacunas de Subunidad/efectos adversos , Vacunas de Subunidad/química , Vacunas de Subunidad/inmunología
18.
Biol Reprod ; 76(2): 218-23, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17050862

RESUMEN

The regulation of early follicular growth and development involves a complex interaction of autocrine, paracrine, and endocrine signals. The ability of these factors to regulate follicle growth may depend in part on the extent of vascular delivery to and perfusion of the ovary. Vascular endothelial growth factor A (VEGFA) is a major regulator of vascular physiology in the ovary. VEGFA is produced in numerous ovarian compartments and likely plays a role in the regulation of all phases of follicular growth, from preantral through preovulatory. The aim of the present study was to further evaluate the role of VEGF in early follicle growth by neutralization of endogenous VEGF or VEGF receptors. Adult mice were injected systemically and prepubertal mice were injected directly under the ovarian bursa with antibodies designed to neutralize VEGF or block interaction with its receptors in the ovary. Both systemic and intrabursal injections of VEGF antibody significantly reduced the number of primordial follicles within 1-3 days after administration without affecting primary or secondary follicle numbers. Primordial follicle numbers were not different from control levels by 30 days after VEGFA antibody administration. Administration of antibodies to the kinase domain receptor (KDR), but not the FMS-like tyrosine receptor (FLT1), for VEGF also resulted in a significant decrease in primordial follicles. These data suggest that VEGF plays a vital role in the maintenance and growth of the primordial follicle pool.


Asunto(s)
Ratones/crecimiento & desarrollo , Ratones/metabolismo , Folículo Ovárico/crecimiento & desarrollo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Anticuerpos/administración & dosificación , Anticuerpos/inmunología , Anticuerpos/farmacología , Ciclo Estral/efectos de los fármacos , Femenino , Inyecciones Intraperitoneales , Folículo Ovárico/efectos de los fármacos , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/inmunología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología
19.
Curr Protein Pept Sci ; 7(2): 137-45, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16611139

RESUMEN

In the past two decades a large initiative has been put forth to understand the biological and pathogenic properties of the human T-cell lymphotropic virus type 1 (HTLV-1); this has ultimately led to the development of various experimental vaccination and therapeutic strategies to combat HTLV-1 infection. The focus of this work is to outline key targets for the design of therapeutics for HTLV-1, such as fusion mediated by the envelope glycoprotein, and to discuss reports of novel vaccines or therapeutics. These strategies include peptide, recombinant protein, DNA, and viral vectors. The final focus of this review is to acquaint the reader with vaccine approaches developed in our laboratory over the last decade. These strategies include the development of envelope glycoprotein derived B-cell epitopes for the induction of neutralizing antibodies, as well as a strategy to generate a multivalent cytotoxic T-lymphocyte (CTL) response against the HTLV-1 Tax antigen.


Asunto(s)
Vacunas contra el SIDA , Infecciones por HTLV-I , Virus Linfotrópico T Tipo 1 Humano/inmunología , Vacunas de Subunidad , Secuencia de Aminoácidos , Animales , Epítopos , Productos del Gen env/química , Productos del Gen env/genética , Productos del Gen env/inmunología , Infecciones por HTLV-I/prevención & control , Infecciones por HTLV-I/terapia , Humanos , Datos de Secuencia Molecular , Péptidos/metabolismo , Péptidos/uso terapéutico , Estructura Secundaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA