Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancer Treat Rev ; 106: 102383, 2022 May.
Article in English | MEDLINE | ID: mdl-35367804

ABSTRACT

Due to the intrinsic genetic instability of tumor cells, aberrant and novel tumor antigens can be expressed and serve as potential targets for cancer immunotherapy. This intrinsic feature can be exploited by cancer immunotherapy, particularly with cancer vaccination. Personalized cancer vaccination strategy can be a potent approach to trigger a broad-based antitumor response that is both beneficial and relevant to individual cancer patients. Also, cancer vaccination strategy can be designed to help elicit immunological memory for long-lasting tumor control. In this review, we describe the different types of personalized cancer vaccines and summarize the completed and ongoing cancer vaccination clinical trials in the last 10 years (database from www.clinicaltrials.gov). We also discuss the pros and cons of using different tumor animal models, i.e. syngeneic models, patient-derived xenografts models and genetically engineered mouse models, as tools for investigating cancer vaccination strategies. Finally, we describe preclinical studies that seek to test new emerging vaccination strategies as well as improving existing methods.


Subject(s)
Cancer Vaccines , Neoplasms , Animals , Antigens, Neoplasm , Cancer Vaccines/therapeutic use , Humans , Immunotherapy/methods , Mice , Neoplasms/drug therapy , Vaccination
2.
Cancers (Basel) ; 13(18)2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34572778

ABSTRACT

As the majority of ovarian cancer (OC) patients are diagnosed with metastatic disease, less than 40% will survive past 5 years after diagnosis. OC is characterized by a succession of remissions and recurrences. The most promising time point for immunotherapeutic interventions in OC is following debulking surgery. Accumulating evidence shows that T cells are important in OC; thus, cancer vaccines capable of eliciting antitumor T cells will be effective in OC treatment. In this review, we discuss different cancer vaccines and propose strategies for their incorporation into the OC standard-of-care regimens. Using the murine ID8 ovarian tumor model, we provide evidence that a cancer vaccine can be effectively combined with OC standard-of-care to achieve greater overall efficacy. We demonstrate several important similarities between the ID8 model and OC patients, in terms of response to immunotherapies, and the ID8 model can be an important tool for evaluating combinatorial regimens and clinical trial designs in OC. Other emerging models, including patient-derived xenograft and genetically engineered mouse models, are continuing to improve and can be useful for evaluating cancer vaccination therapies in the near future. Here, we provide a comprehensive review of the completed and current clinical trials evaluating cancer vaccines in OC.

3.
Front Immunol ; 12: 695150, 2021.
Article in English | MEDLINE | ID: mdl-34149738

ABSTRACT

The emergence of ablative therapies has revolutionized the treatment of inoperable solid tumors. Cryoablation stands out for its uniqueness of operation based on hypothermia, and for its ability to unleash the native tumor antigens, resulting in the generation of anti-tumor immune responses. It is not clearly understood how alterations in the rate of freeze impact the immune response outcomes. In this study, we tested fast freeze and slow freeze rates for their locoregional effectiveness and their ability to elicit immune responses in a B16F10 mouse model of melanoma. Tumor bearing mice treated with fast freeze protocol survived better than the ones treated with slow freeze protocol. Fast freeze resulted in a higher magnitude of CD4+ and CD8+ T-cell responses, and a significantly extended survival post re-challenge. Thus, fast freeze rate should be applied in any future studies employing cryoablation as an in vivo vaccination tool in conjunction with targeted immunotherapies.


Subject(s)
Cryosurgery , Melanoma, Experimental/surgery , Skin Neoplasms/surgery , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cytokines/metabolism , Female , Freezing , Kinetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Myeloid Cells/immunology , Myeloid Cells/metabolism , Necrosis , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Burden
4.
Front Immunol ; 10: 2283, 2019.
Article in English | MEDLINE | ID: mdl-31608067

ABSTRACT

Treatment of solid tumors by ablation techniques has gained momentum in the recent years due to their technical simplicity and reduced morbidity as juxtaposed to surgery. Cryoablation is one of such techniques, known for its uniqueness to destroy the tumors by freezing to lethal temperatures. Freezing the tumor locally and allowing it to remain in situ unleashes an array of tumor antigens to be exposed to the immune system, paving the way for the generation of anti-tumor immune responses. However, the immune responses triggered in most cases are insufficient to eradicate the tumors with systemic spread. Therefore, combination of cryoablation and immunotherapy is a new treatment strategy currently being evaluated for its efficacy, notably in patients with metastatic disease. This article examines the mechanistic fabric of cryoablation for the generation of an effective immune response against the tumors, and various possibilities of its combination with different immunotherapies that are capable of inducing exceptional therapeutic responses. The combinatorial treatment avenues discussed in this article if explored in sufficient profundity, could reach the pinnacle of future cancer medicine.


Subject(s)
Antigens, Neoplasm/immunology , Cryosurgery/methods , Immune System/immunology , Immunotherapy/methods , Neoplasms/therapy , Clinical Trials as Topic , Combined Modality Therapy , Humans , Neoplasms/immunology , Treatment Outcome
5.
Vaccines (Basel) ; 6(4)2018 Nov 23.
Article in English | MEDLINE | ID: mdl-30477198

ABSTRACT

Although different types of therapeutic vaccines against established cancerous lesions in various indications have been developed since the 1990s, their clinical benefit is still very limited. This observed lack of effectiveness in cancer eradication may be partially due to the often deficient immunocompetent status of cancer patients, which may facilitate tumor development by different mechanisms, including immune evasion. The most frequently used cellular vehicle in clinical trials are dendritic cells (DCs), thanks to their crucial role in initiating and directing immune responses. Viable vaccination options using DCs are available, with a positive toxicity profile. For these reasons, despite their limited therapeutic outcomes, DC vaccination is currently considered an additional immunotherapeutic option that still needs to be further explored. In this review, we propose potential actions aimed at improving DC vaccine efficacy by counteracting the detrimental mechanisms recognized to date and implicated in establishing a poor immunocompetent status in cancer patients.

6.
Cancer Treat Rev ; 71: 88-101, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30390423

ABSTRACT

The field of cancer immunotherapy has been revolutionized with the use of immune checkpoint blockade antibodies such as anti-programmed cell death 1 protein (PD-1) and chimeric antigen receptor T cells. Significant clinical benefits are observed in different cancer types with these treatments. While considerable efforts are made in augmenting tumor-specific T cell responses with these therapies, other immunotherapies that actively stimulate endogenous anti-tumor T cells and generating long-term memory have received less attention. Given the high cost of cancer immunotherapies especially with chimeric antigen receptor T cells, not many patients will have access to such treatments. The next-generation of cancer immunotherapy could entail in vivo cancer vaccination to activate both the innate and adaptive anti-tumor responses. This could potentially be achieved via in vivo targeting of dendritic cells which are an indispensable link between the innate and adaptive immunities. Dendritic cells highly expressed toll-like receptors for recognizing and eliminating pathogens. Synthetic toll-like receptors agonists could be synthesized at a low cost and have shown promise in preclinical and clinical trials. As different subsets of human dendritic cells exist in the immune system, activation with different toll-like receptor agonists could exert profound effects on the quality and magnitude of anti-tumor T cell responses. Here, we reviewed the different subsets of human dendritic cells. Using published preclinical and clinical cancers studies available on PubMed, we discussed the use of clinically approved and emerging toll-like receptor agonists to activate dendritic cells in vivo for cancer immunotherapy. Finally, we searched www.clinicaltrials.gov and summarized the active cancer trials evaluating toll-like receptor agonists as an adjuvant.


Subject(s)
Dendritic Cells/immunology , Immunotherapy/methods , Molecular Targeted Therapy/methods , Neoplasms/therapy , Toll-Like Receptors/agonists , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , Cancer Vaccines , Humans , Imiquimod/pharmacology , Langerhans Cells/cytology , Langerhans Cells/drug effects , Langerhans Cells/immunology , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Skin/cytology , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/immunology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/immunology , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/immunology
7.
Vaccines (Basel) ; 3(2): 344-72, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-26343191

ABSTRACT

With its vast amount of uncharacterized and characterized T cell epitopes available for activating CD4⁺ T helper and CD8⁺ cytotoxic lymphocytes simultaneously, whole tumor antigen represents an attractive alternative source of antigens as compared to tumor-derived peptides and full-length recombinant tumor proteins for dendritic cell (DC)-based immunotherapy. Unlike defined tumor-derived peptides and proteins, whole tumor lysate therapy is applicable to all patients regardless of their HLA type. DCs are essentially the master regulators of immune response, and are the most potent antigen-presenting cell population for priming and activating naïve T cells to target tumors. Because of these unique properties, numerous DC-based immunotherapies have been initiated in the clinics. In this review, we describe the different types of whole tumor antigens that we could use to pulse DCs ex vivo and in vivo. We also discuss the different routes of delivering whole tumor antigens to DCs in vivo and activating them with toll-like receptor agonists.

8.
Expert Opin Biol Ther ; 15(4): 569-82, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25553913

ABSTRACT

INTRODUCTION: Dendritic cells (DCs) are the most important antigen-presenting cell population for activating antitumor T-cell responses; therefore, they offer a unique opportunity for specific targeting of tumors. AREAS COVERED: We will discuss the critical factors for the enhancement of DC vaccine efficacy: different DC subsets, types of in vitro DC manufacturing protocol, types of tumor antigen to be loaded and finally different adjuvants for activating them. We will cover potential combinatorial strategies with immunomodulatory therapies: depleting T-regulatory (Treg) cells, blocking VEGF and blocking inhibitory signals. Furthermore, recommendations to incorporate these criteria into DC-based tumor immunotherapy will be suggested. EXPERT OPINION: Monocyte-derived DCs are the most widely used DC subset in the clinic, whereas Langerhans cells and plasmacytoid DCs are two emerging DC subsets that are highly effective in eliciting cytotoxic T lymphocyte responses. Depending on the type of tumor antigens selected for loading DCs, it is important to optimize a protocol that will generate highly potent DCs. The future aim of DC-based immunotherapy is to combine it with one or more immunomodulatory therapies, for example, Treg cell depletion, VEGF blockage and T-cell checkpoint blockage, to elicit the most optimal antitumor immunity to induce long-term remission or even cure cancer patients.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Dendritic Cells/transplantation , Immunotherapy/methods , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell- and Tissue-Based Therapy/trends , Dendritic Cells/immunology , Humans , Immunotherapy/trends , Monocytes/immunology , Monocytes/transplantation , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation
9.
Clin Cancer Res ; 19(17): 4801-15, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23838316

ABSTRACT

PURPOSE: Whole tumor lysates are promising antigen sources for dendritic cell (DC) therapy as they contain many relevant immunogenic epitopes to help prevent tumor escape. Two common methods of tumor lysate preparations are freeze-thaw processing and UVB irradiation to induce necrosis and apoptosis, respectively. Hypochlorous acid (HOCl) oxidation is a new method for inducing primary necrosis and enhancing the immunogenicity of tumor cells. EXPERIMENTAL DESIGN: We compared the ability of DCs to engulf three different tumor lysate preparations, produce T-helper 1 (TH1)-priming cytokines and chemokines, stimulate mixed leukocyte reactions (MLR), and finally elicit T-cell responses capable of controlling tumor growth in vivo. RESULTS: We showed that DCs engulfed HOCl-oxidized lysate most efficiently stimulated robust MLRs, and elicited strong tumor-specific IFN-γ secretions in autologous T cells. These DCs produced the highest levels of TH1-priming cytokines and chemokines, including interleukin (IL)-12. Mice vaccinated with HOCl-oxidized ID8-ova lysate-pulsed DCs developed T-cell responses that effectively controlled tumor growth. Safety, immunogenicity of autologous DCs pulsed with HOCl-oxidized autologous tumor lysate (OCDC vaccine), clinical efficacy, and progression-free survival (PFS) were evaluated in a pilot study of five subjects with recurrent ovarian cancer. OCDC vaccination produced few grade 1 toxicities and elicited potent T-cell responses against known ovarian tumor antigens. Circulating regulatory T cells and serum IL-10 were also reduced. Two subjects experienced durable PFS of 24 months or more after OCDC. CONCLUSIONS: This is the first study showing the potential efficacy of a DC vaccine pulsed with HOCl-oxidized tumor lysate, a novel approach in preparing DC vaccine that is potentially applicable to many cancers.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Lymphocyte Culture Test, Mixed , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Animals , Antigens/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Dendritic Cells/chemistry , Disease-Free Survival , Female , Humans , Hypochlorous Acid/pharmacology , Immunotherapy , Interleukin-10/blood , Mice , Ovarian Neoplasms/chemistry , Ovarian Neoplasms/metabolism , Oxidation-Reduction , T-Lymphocytes, Regulatory/immunology
10.
PLoS One ; 6(12): e28732, 2011.
Article in English | MEDLINE | ID: mdl-22194898

ABSTRACT

"Day-7" myeloid DCs are commonly used in the clinic. However, there is a strong need to develop DCs faster that have the same potent immunostimulatory capacity as "Day-7" myeloid DCs and at the same time minimizing time, labor and cost of DC preparations. Although "2 days" DCs can elicit peptide-specific responses, they have not been demonstrated to engulf, process and present complex whole tumor lysates, which could be more convenient and personalized source of tumor antigens than defined peptides. In this preclinical study, we evaluated the T-cell stimulatory capacity of Day-2, Day-4, and Day-7 cultured monocyte-derived DCs loaded with SKOV3 cell whole lysate prepared by freeze-thaw or by UVB-irradiation followed by freeze-thaw, and matured with lipopolysaccharide (LPS) and interferon (IFN)-gamma. DCs were evaluated for antigen uptake, and following maturation with LPS and IFN-gamma, DCs were assessed for expression of CD80, CD40, CD86, ICAM-1 and CCR7, production of IL-12p70 and IP-10, and induction of tumor-specific T-cell responses. Day-4 and Day-7 DCs exhibited similar phagocytic abilities, which were superior to Day-2 DCs. Mature Day-7 DCs expressed the highest CD40 and ICAM-1, but mature Day-4 DCs produced the most IL-12p70 and IP-10. Importantly, Day-4 and Day-7 DCs derived from ovarian cancer patients stimulated equally strongly tumor-specific T-cell responses. This is the first study demonstrating the highly immunogenic and strong T-cell stimulatory properties of Day-4 myeloid DCs, and provided important preclinical data for rapid development of potent whole tumor lysate-loaded DC vaccines that are applicable to many tumor types.


Subject(s)
Dendritic Cells/immunology , Myeloid Cells/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Tissue Extracts/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Cross-Priming/drug effects , Cytokines/biosynthesis , Dendritic Cells/cytology , Dendritic Cells/drug effects , Female , Health , Humans , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Myeloid Cells/cytology , Myeloid Cells/drug effects , Phagocytosis/drug effects , Phenotype , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tissue Donors
11.
Int Rev Immunol ; 30(2-3): 150-82, 2011.
Article in English | MEDLINE | ID: mdl-21557641

ABSTRACT

Whole tumor cell lysates can serve as excellent multivalent vaccines for priming tumor-specific CD8(+) and CD4(+) T cells. Whole cell vaccines can be prepared with hypochlorous acid oxidation, UVB-irradiation and repeat cycles of freeze and thaw. One major obstacle to successful immunotherapy is breaking self-tolerance to tumor antigens. Clinically approved adjuvants, including Montanide™ ISA-51 and 720, and keyhole-limpet proteins can be used to enhance tumor cell immunogenicity by stimulating both humoral and cellular anti-tumor responses. Other potential adjuvants, such as Toll-like receptor agonists (e.g., CpG, MPLA and PolyI:C), and cytokines (e.g., granulocyte-macrophage colony stimulating factor), have also been investigated.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/immunology , Immunotherapy/methods , Ovarian Neoplasms/immunology , Toll-Like Receptors/agonists , Antibody Formation/immunology , Cancer Vaccines/therapeutic use , Clinical Trials as Topic , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Hemocyanins/administration & dosage , Hemocyanins/immunology , Humans , Mannitol/administration & dosage , Mannitol/analogs & derivatives , Mannitol/immunology , Oleic Acids/administration & dosage , Oleic Acids/immunology , Ovarian Neoplasms/therapy , T-Lymphocytes/immunology , Treatment Outcome
12.
Semin Immunol ; 22(3): 132-43, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20356763

ABSTRACT

Although cancer vaccines with defined antigens are commonly used, the use of whole tumor cell preparations in tumor immunotherapy is a very promising approach and can obviate some important limitations in vaccine development. Whole tumor cells are a good source of TAAs and can induce simultaneous CTLs and CD4(+) T helper cell activation. We review current approaches to prepare whole tumor cell vaccines, including traditional methods of freeze-thaw lysates, tumor cells treated with ultraviolet irradiation, and RNA electroporation, along with more recent methods to increase tumor cell immunogenicity with HOCl oxidation or infection with replication-incompetent herpes simplex virus.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Dendritic Cells/cytology , Ovarian Neoplasms/immunology , Animals , Apoptosis , Female , Humans , Immunotherapy/methods , Mice , Necrosis , Ovarian Neoplasms/therapy , Simplexvirus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...