Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Nanomedicine ; 62: 102776, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39102973

RESUMEN

Selective in vivo immune cell manipulation offers a promising strategy for cancer vaccines. In this context, spatiotemporal control over recruitment of specific cells, and their direct exposure to appropriate immunoadjuvants and antigens are key to effective cancer vaccines. We present an implantable 3D-printed cancer vaccine platform called the 'NanoLymph' that enables spatiotemporally-controlled recruitment and manipulation of immune cells in a subcutaneous site. Leveraging two reservoirs each for continuous immunoadjuvant release or antigen presentation, the NanoLymph attracts dendritic cells (DCs) on site and exposes them to tumor-associated antigens. Upon local antigen-specific activation, DCs are mobilized to initiate a systemic immune response. NanoLymph releasing granulocyte-macrophage colony-stimulating factor and CpG-oligodeoxynucleotides with irradiated whole cell tumor lysate inhibited tumor growth of B16F10 murine melanoma in a prophylactic and therapeutic vaccine setting. Overall, this study presents the NanoLymph as a versatile cancer vaccine development platform with replenishable and controlled local release of antigens and immunoadjuvants.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas , Inmunomodulación , Melanoma Experimental , Animales , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/administración & dosificación , Ratones , Células Dendríticas/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Adyuvantes Inmunológicos , Femenino , Oligodesoxirribonucleótidos/inmunología , Humanos
2.
Curr Issues Mol Biol ; 45(11): 9215-9233, 2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-37998754

RESUMEN

The treatment of unresectable or metastatic Head and Neck Squamous Cell Carcinoma (HNSCC) has traditionally relied on chemotherapy or radiotherapy, yielding suboptimal outcomes. The introduction of immunotherapy has significantly improved HNSCC treatment, even if the long-term results cannot be defined as satisfactory. Its mechanism of action aims to counteract the blockade of tumor immune escape. This result can also be obtained by stimulating the immune system with vaccines. This review scope is to comprehensively gather existing evidence and summarize ongoing clinical trials focused on therapeutic vaccines for HNSCC treatment. The current landscape reveals numerous promising drugs in the early stages of experimentation, along with a multitude of trials that have been suspended or abandoned for years. Nonetheless, there are encouraging results and ongoing experiments that instill hope for potential paradigm shifts in HNSCC therapy.

3.
Curr Treat Options Oncol ; 24(5): 496-514, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36976454

RESUMEN

OPINION STATEMENT: The development of immunotherapies for nonmelanoma skin cancer (NMSC) has lagged far behind that for melanoma in the past few decades, given that the majority of cases are surgically curable. Nevertheless, given the steady growth in the incidence rate of NMSC and attendant increase in patients with unresectable or advanced-stage tumors, the demand for systemic therapy is noticeably increasing. To date, the most widely used immunotherapeutic strategies, including immune checkpoint inhibitors and T-cell therapy, have obtained satisfactory results in some patients but not others. Even with an objective response in a fraction of patients, some accompanying adverse events may lead to intolerance and noncompliance. The expanding understanding of immune surveillance and tumor escape has provided us with novel perspectives in the field of immunotherapy. One emerging approach, the therapeutic cancer vaccine, encompasses the potential to newly "prime" T cells by activating antigen presentation in regional lymph nodes and the tumor microenvironment. Immune cells are therefore preconditioned and awakened to be ready to attack tumors. In NMSCs, multiple clinical trials of cancer vaccines are underway. The vaccine targets include tumor-associated antigens, tumor-specific antigens, oncolytic viruses, and toll-like receptors. Although clinical benefits have been shown in specific case reports and trials, various challenges remain to be resolved to guarantee applicability in the general patient population. Standing on the shoulders of pioneers expedites the pace of advances in therapeutic cancer vaccines, making them the rising star in the field of immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Melanoma , Neoplasias Cutáneas , Humanos , Vacunas contra el Cáncer/uso terapéutico , Neoplasias Cutáneas/terapia , Antígenos de Neoplasias/uso terapéutico , Inmunoterapia/métodos , Microambiente Tumoral
4.
Int J Mol Sci ; 24(3)2023 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-36768214

RESUMEN

The purpose of immune checkpoint inhibitor (ICI)-based therapies is to help the patient's immune system to combat tumors by restoring the immune response mediated by CD8+ cytotoxic T cells. Despite impressive clinical responses, most patients do not respond to ICIs. Therapeutic vaccines with autologous professional antigen-presenting cells, including dendritic cells, do not show yet significant clinical benefit. To improve these approaches, we have developed a new therapeutic vaccine based on an allogeneic plasmacytoid dendritic cell line (PDC*line), which efficiently activates the CD8+ T-cell response in the context of melanoma. The goal of the study is to demonstrate the potential of this platform to activate circulating tumor-specific CD8+ T cells in patients with lung cancer, specifically non-small-cell lung cancer (NSCLC). PDC*line cells loaded with peptides derived from tumor antigens are used to stimulate the peripheral blood mononuclear cells of NSCLC patients. Very interestingly, we demonstrate an efficient activation of specific T cells for at least two tumor antigens in 69% of patients irrespective of tumor antigen mRNA overexpression and NSCLC subtype. We also show, for the first time, that the antitumor CD8+ T-cell expansion is considerably improved by clinical-grade anti-PD-1 antibodies. Using PDC*line cells as an antigen presentation platform, we show that circulating antitumor CD8+ T cells from lung cancer patients can be activated, and we demonstrate the synergistic effect of anti-PD-1 on this expansion. These results are encouraging for the development of a PDC*line-based vaccine in NSCLC patients, especially in combination with ICIs.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Leucocitos Mononucleares/patología , Linfocitos T CD8-positivos , Antígenos de Neoplasias , Células Dendríticas
5.
Mol Cancer ; 21(1): 102, 2022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-35459256

RESUMEN

BACKGROUND: Redirecting pre-existing virus-specific cytotoxic CD8+ T lymphocytes (CTLs) to tumors by simulating a viral infection of the tumor cells has great potential for cancer immunotherapy. However, this strategy is limited by lack of amenable method for viral antigen delivery into the cytosol of target tumors. Here, we addressed the limit by developing a CD8+ T cell epitope-delivering antibody, termed a TEDbody, which was engineered to deliver a viral MHC-I epitope peptide into the cytosol of target tumor cells by fusion with a tumor-specific cytosol-penetrating antibody. METHODS: To direct human cytomegalovirus (CMV)-specific CTLs against tumors, we designed a series of TEDbodies carrying various CMV pp65 antigen-derived peptides. CMV-specific CTLs from blood of CMV-seropositive healthy donors were expanded for use in in vitro and in vivo experiments. Comprehensive cellular assays were performed to determine the presentation mechanism of TEDbody-mediated CMV peptide-MHC-I complex (CMV-pMHCI) on the surface of target tumor cells and the recognition and lysis by CMV-specific CTLs. In vivo CMV-pMHCI presentation and antitumor efficacy of TEDbody were evaluated in immunodeficient mice bearing human tumors. RESULTS: TEDbody delivered the fused epitope peptides into target tumor cells to be intracellularly processed and surface displayed in the form of CMV-pMHCI, leading to disguise target tumor cells as virally infected cells for recognition and lysis by CMV-specific CTLs. When systemically injected into tumor-bearing immunodeficient mice, TEDbody efficiently marked tumor cells with CMV-pMHCI to augment the proliferation and cytotoxic property of tumor-infiltrated CMV-specific CTLs, resulting in significant inhibition of the in vivo tumor growth by redirecting adoptively transferred CMV-specific CTLs. Further, combination of TEDbody with anti-OX40 agonistic antibody substantially enhanced the in vivo antitumor activity. CONCLUSION: Our study offers an effective technology for MHC-I antigen cytosolic delivery. TEDbody may thus have utility as a therapeutic cancer vaccine to redirect pre-existing anti-viral CTLs arising from previously exposed viral infections to attack tumors.


Asunto(s)
Infecciones por Citomegalovirus , Neoplasias , Animales , Linfocitos T CD8-positivos , Infecciones por Citomegalovirus/terapia , Citosol , Epítopos , Humanos , Inmunoterapia/métodos , Ratones , Péptidos , Linfocitos T Citotóxicos
6.
J Transl Med ; 20(1): 419, 2022 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-36089578

RESUMEN

BACKGROUND: This clinical trial evaluated a novel telomerase-targeting therapeutic cancer vaccine, UV1, in combination with ipilimumab, in patients with metastatic melanoma. Translational research was conducted on patient-derived blood and tissue samples with the goal of elucidating the effects of treatment on the T cell receptor repertoire and tumor microenvironment. METHODS: The trial was an open-label, single-center phase I/IIa study. Eligible patients had unresectable metastatic melanoma. Patients received up to 9 UV1 vaccinations and four ipilimumab infusions. Clinical responses were assessed according to RECIST 1.1. Patients were followed up for progression-free survival (PFS) and overall survival (OS). Whole-exome and RNA sequencing, and multiplex immunofluorescence were performed on the biopsies. T cell receptor (TCR) sequencing was performed on the peripheral blood and tumor tissues. RESULTS: Twelve patients were enrolled in the study. Vaccine-specific immune responses were detected in 91% of evaluable patients. Clinical responses were observed in four patients. The mPFS was 6.7 months, and the mOS was 66.3 months. There was no association between baseline tumor mutational burden, neoantigen load, IFN-γ gene signature, tumor-infiltrating lymphocytes, and response to therapy. Tumor telomerase expression was confirmed in all available biopsies. Vaccine-enriched TCR clones were detected in blood and biopsy, and an increase in the tumor IFN-γ gene signature was detected in clinically responding patients. CONCLUSION: Clinical responses were observed irrespective of established predictive biomarkers for checkpoint inhibitor efficacy, indicating an added benefit of the vaccine-induced T cells. The clinical and immunological read-out warrants further investigation of UV1 in combination with checkpoint inhibitors. Trial registration Clinicaltrials.gov identifier: NCT02275416. Registered October 27, 2014. https://clinicaltrials.gov/ct2/show/NCT02275416?term=uv1&draw=2&rank=6.


Asunto(s)
Melanoma , Telomerasa , Humanos , Ipilimumab/farmacología , Ipilimumab/uso terapéutico , Melanoma/patología , Microambiente Tumoral , Vacunación
7.
Mol Ther ; 29(2): 555-570, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33038322

RESUMEN

Tremendous innovation is underway among a rapidly expanding repertoire of promising personalized immune-based treatments. Therapeutic cancer vaccines (TCVs) are attractive systemic immunotherapies that activate and expand antigen-specific CD8+ and CD4+ T cells to enhance anti-tumor immunity. Our review highlights key issues impacting TCVs in clinical practice and reports on progress in development. We review the mechanism of action, immune-monitoring, dosing strategies, combinations, obstacles, and regulation of cancer vaccines. Most trials of personalized TCVs are ongoing and represent diverse platforms with predominantly early investigations of mRNA, DNA, or peptide-based targeting strategies against neoantigens in solid tumors, with many in combination immunotherapies. Multiple delivery systems, routes of administration, and dosing strategies are used. Intravenous or intramuscular administration is common, including delivery by lipid nanoparticles. Absorption and biodistribution impact antigen uptake, expression, and presentation, affecting the strength, speed, and duration of immune response. The emerging trials illustrate the complexity of developing this class of innovative immunotherapies. Methodical testing of the multiple potential factors influencing immune responses, as well as refined quantitative methodologies to facilitate optimal dosing strategies, could help resolve uncertainty of therapeutic approaches. To increase the likelihood of success in bringing these medicines to patients, several unique development challenges must be overcome.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor , Ensayos Clínicos como Asunto , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Desarrollo de Medicamentos , Humanos , Inmunoterapia/métodos , Medicina de Precisión/métodos , Linfocitos T/inmunología
8.
J Liposome Res ; 32(1): 22-31, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33006297

RESUMEN

Cancer vaccines have been developed as an additional method of treatment in the fight against cancer. However, an important barrier to an effective vaccine is the inefficient presentation of exogenous antigen by dendritic cells to cytotoxic CD8 T cells. In this study, DPPC liposomes were modified with channels and loaded with polyethyleneimine (PEI) and 5,6-dimethylxanthenone-4-acetic acid (DMXAA) to produce a vaccine carrier. The liposomes were designed to be pH responsive to facilitate delivery of antigens directly to the cytoplasm of antigen presenting cells, bypassing the cross-presentation pathway and improving cellular immune responses. The lysis of liposomes in acidic cell-free conditions was measured using a validated dynamic light scattering assay in order to gain an insight into the mechanism of PEI-mediated lysis. Dendritic cell stimulation and T cell proliferation was investigated in vitro and the potential of this formulation to stimulate a therapeutic anti-cancer immune response was examined in a murine melanoma model. The modified formulation stimulated T cell activation in vitro and induced a small but significant increase in survival in immunized mice. Overall, liposomes modified with PEI and channels successfully delivered antigen to the cytoplasm of dendritic cells, which subsequently led to the development of an appropriate immune response.


Asunto(s)
Ciclodextrinas , Nanopartículas , Vacunas , Animales , Citoplasma , Células Dendríticas , Liposomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Polietileneimina
9.
Int J Mol Sci ; 22(15)2021 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-34360800

RESUMEN

Therapeutic cancer vaccines have become increasingly qualified for use in personalized cancer immunotherapy. A deeper understanding of tumor immunology and novel antigen delivery technologies has assisted in optimizing vaccine design. Therapeutic cancer vaccines aim to establish long-lasting immunological memory against tumor cells, thereby leading to effective tumor regression and minimizing non-specific or adverse events. However, due to several resistance mechanisms, significant challenges remain to be solved in order to achieve these goals. In this review, we describe our current understanding with respect to the use of the antigen repertoire in vaccine platform development. We also summarize various intrinsic and extrinsic resistance mechanisms behind the failure of cancer vaccine development in the past. Finally, we suggest a strategy that combines immune checkpoint inhibitors to enhance the efficacy of cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia , Neoplasias/terapia , Medicina de Precisión , Animales , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/uso terapéutico , Humanos , Neoplasias/inmunología
10.
Pharm Res ; 35(11): 207, 2018 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-30209623

RESUMEN

PURPOSE: Personalized peptide-based cancer vaccines will be composed of multiple patient specific synthetic long peptides (SLPs) which may have various physicochemical properties. To formulate such SLPs, a flexible vaccine delivery system is required. We studied whether cationic liposomes are suitable for this purpose. METHODS: Fifteen SIINFEKL T cell epitope-containing SLPs, widely differing in hydrophobicity and isoelectric point, were separately loaded in cationic liposomes via the dehydration-rehydration method. Particle size and polydispersity index (PDI) were measured via dynamic light scattering (DLS), and zeta potential with laser Doppler electrophoresis. Peptide loading was fluorescently determined and the immunogenicity of the formulated peptides was assessed in co-cultures of dendritic cells (DCs) and CD8+ T-cells in vitro. RESULTS: All SLPs were loaded in cationic liposomes by using three different loading method variants, depending on the SLP characteristics. The fifteen liposomal formulations had a comparable size (< 200 nm), PDI (< 0.3) and zeta potential (22-30 mV). Cationic liposomes efficiently delivered the SLPs to DCs that subsequently activated SIINFEKL-specific CD8+ T-cells, indicating improved immunological activity of the SLPs. CONCLUSION: Cationic liposomes can accommodate a wide range of different SLPs and are therefore a potential delivery platform for personalized cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Portadores de Fármacos/química , Epítopos de Linfocito T , Liposomas/química , Oligopéptidos/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Cationes , Composición de Medicamentos , Liberación de Fármacos , Colorantes Fluorescentes/química , Humanos , Activación de Linfocitos , Oligopéptidos/química , Oligopéptidos/inmunología , Ovalbúmina/química , Tamaño de la Partícula , Fragmentos de Péptidos/química , Biblioteca de Péptidos , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/química , Vacunas de Subunidad/inmunología
11.
Mol Pharm ; 14(5): 1482-1493, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28088862

RESUMEN

The aim of the present study was to improve the immunogenicity of peptide epitope vaccines using novel nanocarriers based on self-assembling materials. Several studies demonstrated that peptide antigens in nanoparticulate form induce stronger immune responses than their soluble forms. However, several issues such as poor loading and risk of inducing T cell anergy due to premature release of antigenic epitopes have challenged the clinical success of such systems. In the present study, we developed two vaccine delivery systems by appending a self-assembling peptide (Ac-AAVVLLLW-COOH) or a thermosensitive polymer poly(N-isopropylacrylamide (pNIPAm) to the N-terminus of different peptide antigens (OVA250-264, HPV-E743-57) to generate self-assembling peptide epitopes (SAPEs). The obtained results showed that the SAPEs were able to form nanostructures with a diameter from 20 to 200 nm. The SAPEs adjuvanted with CpG induced and expanded antigen-specific CD8+ T cells in mice. Furthermore, tumor-bearing mice vaccinated with SAPEs harboring the HPV E743-57 peptide showed a delayed tumor growth and an increased survival compared to sham-treated mice. In conclusion, self-assembling peptide based systems increase the immunogenicity of peptide epitope vaccines and therefore warrants further development toward clinical use.


Asunto(s)
Epítopos de Linfocito T/metabolismo , Péptidos/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/metabolismo , Células Cultivadas , Epítopos de Linfocito T/inmunología , Inmunoterapia , Ratones , Nanopartículas/química , Ovalbúmina/química , Vacunación/métodos
12.
Stat Med ; 36(4): 592-605, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-27807870

RESUMEN

Arming the immune system against cancer has emerged as a powerful tool in oncology during recent years. Instead of poisoning a tumor or destroying it with radiation, therapeutic cancer vaccine, a type of cancer immunotherapy, unleashes the immune system to combat cancer. This indirect mechanism-of-action of vaccines poses the possibility of a delayed onset of clinical effect, which results in a delayed separation of survival curves between the experimental and control groups in therapeutic cancer vaccine trials with time-to-event endpoints. This violates the proportional hazard assumption. As a result, the conventional study design based on the regular log-rank test ignoring the delayed effect would lead to a loss of power. In this paper, we propose two innovative approaches for sample size and power calculation using the piecewise weighted log-rank test to properly and efficiently incorporate the delayed effect into the study design. Both theoretical derivations and empirical studies demonstrate that the proposed methods, accounting for the delayed effect, can reduce sample size dramatically while achieving the target power relative to a standard practice. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Neoplasias/terapia , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos , Estadística como Asunto/métodos , Interpretación Estadística de Datos , Humanos , Estimación de Kaplan-Meier , Modelos Estadísticos , Neoplasias/inmunología , Neoplasias/mortalidad , Modelos de Riesgos Proporcionales , Análisis de Supervivencia , Factores de Tiempo
13.
Bioorg Med Chem ; 24(18): 4372-4380, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27475535

RESUMEN

Immunotherapy is one of the most promising strategies for the treatment of cancer. Human papillomavirus (HPV) is responsible for virtually all cases of cervical cancer. The main purpose of a therapeutic HPV vaccine is to stimulate CD8(+) cytotoxic T lymphocytes (CTLs) that can eradicate HPV infected cells. HPV oncoproteins E6 and E7 are continuously expressed and are essential for maintaining the growth of HPV-associated tumor cells. We designed polymer-based multi-antigenic formulations/constructs that were comprised of the E6 and E7 peptide epitopes. We developed an N-terminus-based epitope conjugation to conjugate two unprotected peptides to poly tert-butyl acrylate. This method allowed for the incorporation of the two antigens into a polymeric dendrimer in a strictly equimolar ratio. The most effective formulations eliminated tumors in up to 50% of treated mice. Tumor recurrence was not observed up to 3months post initial challenge.


Asunto(s)
Antígenos/química , Vacunas contra Papillomavirus/uso terapéutico , Péptidos/química , Polímeros/química , Neoplasias del Cuello Uterino/prevención & control , Secuencia de Aminoácidos , Animales , Cromatografía Líquida de Alta Presión , Epítopos/química , Epítopos/inmunología , Femenino , Ratones , Ratones Endogámicos C57BL , Vacunas contra Papillomavirus/química , Vacunas contra Papillomavirus/inmunología , Espectrometría de Masa por Ionización de Electrospray
14.
Adv Exp Med Biol ; 909: 139-67, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27240458

RESUMEN

Cancer is one of the major leading death causes of diseases. Prevention and treatment of cancer is an important way to decrease the incidence of tumorigenesis and prolong patients' lives. Subversive achievements on cancer immunotherapy have recently been paid much attention after many failures in basic and clinical researches. Based on deep analysis of genomics and proteomics of tumor antigens, a variety of cancer vaccines targeting tumor antigens have been tested in preclinical and human clinical trials. Many therapeutic cancer vaccines alone or combination with other conventional treatments for cancer obtained spectacular efficacy, indicating the tremendously potential application in clinic. With the illustration of underlying mechanisms of cancer immune regulation, valid, controllable, and persistent cancer vaccines will play important roles in cancer treatment, survival extension and relapse and cancer prevention. This chapter mainly summarizes the recent progresses and developments on cancer vaccine research and clinical application, thus exploring the existing obstacles in cancer vaccine research and promoting the efficacy of cancer vaccine.


Asunto(s)
Antineoplásicos/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Terapia Combinada/métodos , Regulación Neoplásica de la Expresión Génica/inmunología , Factores Inmunológicos/uso terapéutico , Neoplasias/terapia , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Humanos , Inmunoterapia/métodos , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/inmunología , Transducción de Señal , Análisis de Supervivencia
15.
BJU Int ; 116(3): 321-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24612369

RESUMEN

The principal role of the immune system is to prevent and eradicate pathogens and infections. The key characteristics or features of an effective immune response include specificity, trafficking, antigen spread and durability (memory). The immune system is recognised to have a critical role in controlling cancer through a dynamic relationship with tumour cells. Normally, at the early stages of tumour development, the immune system is capable of eliminating tumour cells or keeping tumour growth abated; however, tumour cells may evolve multiple pathways over time to evade immune control. Immunotherapy may be viewed as a treatment designed to boost or restore the ability of the immune system to fight cancer, infections and other diseases. Immunotherapy manifests differently from traditional cancer treatments, eliciting delayed response kinetics and thus may be more effective in patients with lower tumour burden, in whom disease progression may be less rapid, thereby allowing ample time for the immunotherapy to evolve. Because immunotherapies may have a different mechanism of action from traditional cytotoxic or targeted biological agents, immunotherapy techniques have the potential to combine synergistically with traditional therapies.


Asunto(s)
Inmunoterapia , Neoplasias , Anticuerpos Monoclonales/uso terapéutico , Vacunas contra el Cáncer , Citocinas , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología
16.
Crit Rev Oncol Hematol ; 193: 104198, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37949152

RESUMEN

Therapeutic cancer vaccines have shown promising efficacy in helping immunotherapy for cancer patients, but the systematic characterization of the clinical application and the method for improving efficacy is lacking. Here, we mainly summarize the classification of therapeutic cancer vaccines, including protein vaccines, nucleic acid vaccines, cellular vaccines and anti-idiotypic antibody vaccines, and subdivide the above vaccines according to different types and delivery forms. Additionally, we outline the clinical efficacy and safety of vaccines, as well as the combination strategies of therapeutic cancer vaccines with other therapies. This review will provide a detailed overview and rationale for the future clinical application and development of therapeutic cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Humanos , Vacunas contra el Cáncer/uso terapéutico , Neoplasias/tratamiento farmacológico , Inmunoterapia/métodos , Proteínas , Resultado del Tratamiento
17.
Adv Sci (Weinh) ; 11(36): e2403663, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39073756

RESUMEN

Clinical translation of current cancer vaccine research has been hampered by limited antitumor immune responses due to inefficient antigen delivery and presentation, suboptimal DC and T cell activation. Biomaterial-based nanovaccine offers targeted antigen delivery, protection from degradation in vivo, and prolonged tumor therapeutic efficacy. This study introduces a lipid-coated deoxycholic acid-survivin nanoassembly (DA-L-DSA). Survivin, overexpressed in several cancer cells and involved in cancer cell growth and immune evasion, is selected as a tumor-associated antigen. An major histocompatibility complex class I binding epitope of survivin is engineered into the nanoassembly. R848, TLR 7/8 agonist, and SD-208, TGF-beta receptor1 kinase inhibitor, are coencapsulated into the nanoassembly as potent adjuvants to boost DC maturation and enhance antigen presentation. The DA-L-DSA effectively stimulates the maturation of dendritic cells, migrates into lymph nodes, and enhances T-cell activation and Th1 response. A substantial influx of cytotoxic T lymphocytes into primary tumors is observed in a murine melanoma model and demonstrates anti-metastatic effects in a spontaneous breast cancer metastasis model. Furthermore, DA-L-DSA exhibits a remarkable synergistic effect in the combination therapy with immune checkpoint inhibitors alleviating immunosuppressive tumor microenvironment. Taken together, these findings suggest DA-L-DSA as a promising immuno-therapeutic platform that could be applicable to diverse intractable cancers.


Asunto(s)
Adyuvantes Inmunológicos , Vacunas contra el Cáncer , Células Dendríticas , Inmunoterapia , Animales , Células Dendríticas/inmunología , Ratones , Inmunoterapia/métodos , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/administración & dosificación , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Survivin/inmunología , Antígenos de Neoplasias/inmunología , Femenino , Humanos , Péptidos/inmunología , Péptidos/química
18.
Hum Vaccin Immunother ; 20(1): 2379864, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-39165083

RESUMEN

This Phase I/IIa open-label, single-arm clinical trial addressing advanced, refractory, metastatic breast cancer was conducted at six medical centers in the United States. We repeated inoculations with irradiated SV-BR-1-GM, a breast cancer cell line with antigen-presenting activity engineered to release granulocyte-macrophage colony-stimulating factor (GM-CSF), with pre-dose low-dose cyclophosphamide and post-dose local interferon alpha. Twenty-six patients were enrolled; 23 (88.5%) were inoculated, receiving a total of 79 inoculations. There were six Grade 4 and one Grade 5 adverse events noted (judged unrelated to SV-BR-1-GM). Disease control (stable disease [SD]) occurred in 8 of 16 evaluable patients; 4 showed objective regression of metastases, including 1 patient with near-complete regressions in 20 of 20 pulmonary lesions. All patients with regressions had human leukocyte antigen (HLA) matches with SV-BR-1-GM; non-responders were equally divided between matching and nonmatching (p = .01, Chi-squared), and having ≥2 HLA matches with SV-BR-1-GM (n = 6) correlated with clinical benefit. Delayed-type hypersensitivity (DTH) testing to candida antigen and SV-BR-1-GM generated positive responses (≥5 mm) in 11 (42.3%) and 13 (50%) patients, respectively. Quantifying peripheral circulating tumor cells (CTCs) and cancer-associated macrophage-like cells (CAMLs) showed that a drop in CAMLs was significantly correlated with an improvement in progression-free survival (PFS; 4.1 months vs. 1.8 months, p = .0058). Eight of 10 patients significantly upregulated programmed cell death ligand 1 (PD-L1) on CTCs/CAMLs with treatment (p = .0012). These observations support the safety of the Bria-IMT regimen, demonstrate clinical regressions, imply a role for HLA matching, and identify a possible value for monitoring CAMLs in peripheral blood.


Asunto(s)
Neoplasias de la Mama , Ciclofosfamida , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Interferón-alfa , Humanos , Femenino , Persona de Mediana Edad , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Ciclofosfamida/administración & dosificación , Ciclofosfamida/uso terapéutico , Adulto , Anciano , Interferón-alfa/administración & dosificación , Interferón-alfa/uso terapéutico , Metástasis de la Neoplasia , Línea Celular Tumoral , Resultado del Tratamiento , Estados Unidos
19.
Int Immunopharmacol ; 126: 111225, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37988911

RESUMEN

Therapeutic cancer vaccines are novel immuno-therapeutics, aiming to improve clinical outcomes with other immunotherapies. However, obstacles to their successful clinical development remain, which model-informed drug development approaches may address. UV1 is a telomerase based therapeutic cancer vaccine candidate being investigated in phase I clinical trials for multiple indications. We developed a mechanism-based model structure, using a nonlinear mixed-effects modeling techniques, based on longitudinal tumor sizes (sum of the longest diameters, SLD), UV1-specific immunological assessment (stimulation index, SI) and overall survival (OS) data obtained from a UV1 phase I trial including non-small cell lung cancer (NSCLC) patients and a phase I/IIa trial including malignant melanoma (MM) patients. The final structure comprised a mechanistic tumor growth dynamics (TGD) model, a model describing the probability of observing a UV1-specific immune response (SI ≥ 3) and a time-to-event model for OS. The mechanistic TGD model accounted for the interplay between the vaccine peptides, immune system and tumor. The model-predicted UV1-specific effector CD4+ T cells induced tumor shrinkage with half-lives of 103 and 154 days in NSCLC and MM patients, respectively. The probability of observing a UV1-specific immune response was mainly driven by the model-predicted UV1-specific effector and memory CD4+ T cells. A high baseline SLD and a high relative increase from nadir were identified as main predictors for a reduced OS in NSCLC and MM patients, respectively. Our model predictions highlighted that additional maintenance doses, i.e. UV1 administration for longer periods, may result in more sustained tumor size shrinkage.


Asunto(s)
Vacunas contra el Cáncer , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Melanoma , Telomerasa , Humanos , Vacunas contra el Cáncer/uso terapéutico , Telomerasa/uso terapéutico , Neoplasias Pulmonares/patología , Péptidos/uso terapéutico
20.
Fundam Res ; 3(6): 1005-1024, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38933006

RESUMEN

Immunotherapy has rejuvenated cancer therapy, especially after anti-PD-(L)1 came onto the scene. Among the many therapeutic options, therapeutic cancer vaccines are one of the most essential players. Although great progress has been made in research on tumor antigen vaccines, few phase III trials have shown clinical benefits. One of the reasons lies in obstruction from the tumor microenvironment (TME). Meanwhile, the therapeutic cancer vaccine reshapes the TME in an ambivalent way, leading to immune stimulation or immune escape. In this review, we summarize recent progress on the interaction between therapeutic cancer vaccines and the TME. With respect to vaccine resistance, innate immunosuppressive TME components and acquired resistance caused by vaccination are both involved. Understanding the underlying mechanism of this crosstalk provides insight into the treatment of cancer by directly targeting the TME or synergizing with other therapeutics.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda