Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
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
2.
Nat Nanotechnol ; 15(12): 1043-1052, 2020 12.
Article in English | MEDLINE | ID: mdl-33139933

ABSTRACT

Cancer metastases and recurrence after surgical resection remain an important cause of treatment failure. Here we demonstrate a general strategy to fabricate personalized nanovaccines based on a cationic fluoropolymer for post-surgical cancer immunotherapy. Nanoparticles formed by mixing the fluoropolymer with a model antigen ovalbumin, induce dendritic cell maturation via the Toll-like receptor 4 (TLR4)-mediated signalling pathway, and promote antigen transportation into the cytosol of dendritic cells, which leads to an effective antigen cross-presentation. Such a nanovaccine inhibits established ovalbumin-expressing B16-OVA melanoma. More importantly, a mix of the fluoropolymer with cell membranes from resected autologous primary tumours synergizes with checkpoint blockade therapy to inhibit post-surgical tumour recurrence and metastases in two subcutaneous tumour models and an orthotopic breast cancer tumour. Furthermore, in the orthotopic tumour model, we observed a strong immune memory against tumour rechallenge. Our work offers a simple and general strategy for the preparation of personalized cancer vaccines to prevent post-operative cancer recurrence and metastasis.


Subject(s)
Cancer Vaccines/therapeutic use , Fluorocarbon Polymers/therapeutic use , Melanoma, Experimental/prevention & control , Nanoparticles/therapeutic use , Animals , Cancer Vaccines/chemistry , Cells, Cultured , Female , Fluorocarbon Polymers/chemistry , Immunotherapy , Melanoma, Experimental/immunology , Melanoma, Experimental/surgery , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/chemistry , Ovalbumin/therapeutic use , Polyethyleneimine/chemistry , Polyethyleneimine/therapeutic use
3.
Biomed Pharmacother ; 128: 110294, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32485571

ABSTRACT

In this study, the antitumor and immunomodulatory effects of mesenchymal stem cells (MSC) obtained from bone marrow in the treatment of dorsal melanoma B16-F10. The MSC cells were obtained from the bone marrow of isogenic C57BL/6J mice, characterized and inoculated by two routes, intratumor (it) and intravenous (iv). The hematological profile, expression markers and receptors, phases of the cell cycle and mitochondrial electrical potential were evaluated by flow cytometry. The dorsal tumor mass showed a significant reduction after treatment by the two routes of administration with a significant effect by the intravenous route. MSC showed immunomodulatory potential and did not induce an increase in the markers involved in tumor control and progression. The number of cells in the sub-G1 phase increased significantly after treatments compared to the control group. The percentage of cells in phases G0/G1, S and G2/M decreased, with only the group (it) showing a significant reduction. The intratumor group showed a significant decrease in the G2/M phase. Treatment with MSC provided a significant decrease in the percentage of metabolically active tumor cells, demonstrating its intrinsic effect in the control of cell proliferation. Regarding the mechanism of cell death, MSCs modulated the expression of proteins involved in the regulation of the cell cycle, angiogenesis receptors and pro-apoptotic proteins by intrinsic and extrinsic routes. Therefore, the use of undifferentiated MSC, administered intratumor and intravenous is possibly a promising treatment for melanoma.


Subject(s)
Melanoma, Experimental/surgery , Mesenchymal Stem Cell Transplantation , Angiogenic Proteins/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Tumor Burden
4.
Sci Adv ; 5(10): eaaw6870, 2019 10.
Article in English | MEDLINE | ID: mdl-31681841

ABSTRACT

Erythrocytes or red blood cells (RBCs) represent a promising cell-mediated drug delivery platform due to their inherent biocompatibility. Here, we developed an antigen delivery system based on the nanoerythrosomes derived from RBCs, inspired by the splenic antigen-presenting cell targeting capacity of senescent RBCs. Tumor antigens were loaded onto the nanoerythrosomes by fusing tumor cell membrane-associated antigens with nanoerythrosomes. This tumor antigen-loaded nanoerythrosomes (nano-Ag@erythrosome) elicited antigen responses in vivo and, in combination with the anti-programmed death ligand 1 (PD-L1) blockade, inhibited the tumor growth in B16F10 and 4T1 tumor models. We also generated a tumor model showing that "personalized nano-Ag@erythrosomes" could be achieved by fusing RBCs and surgically removed tumors, which effectively reduced tumor recurrence and metastasis after surgery.


Subject(s)
Antigens/immunology , Erythrocytes/metabolism , Immunotherapy , Nanoparticles/chemistry , Neoplasms/immunology , Neoplasms/therapy , Proteolipids/chemistry , Animals , Antigen-Presenting Cells/metabolism , B7-H1 Antigen/metabolism , Biomarkers/metabolism , Dendritic Cells/metabolism , Macrophages/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/surgery , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasm Recurrence, Local/pathology , Spleen/pathology , Treatment Outcome
5.
Sci Rep ; 6: 35176, 2016 10 12.
Article in English | MEDLINE | ID: mdl-27731390

ABSTRACT

Since its first implementation in otolaryngological surgery nearly a century ago, the surgical microscope has improved the accuracy and the safety of microsurgeries. However, the microscope shows only a magnified surface view of the surgical region. To overcome this limitation, either optical coherence tomography (OCT) or photoacoustic microscopy (PAM) has been independently combined with conventional surgical microscope. Herein, we present a near-infrared virtual intraoperative photoacoustic optical coherence tomography (NIR-VISPAOCT) system that combines both PAM and OCT with a conventional surgical microscope. Using optical scattering and absorption, the NIR-VISPAOCT system simultaneously provides surgeons with real-time comprehensive biological information such as tumor margins, tissue structure, and a magnified view of the region of interest. Moreover, by utilizing a miniaturized beam projector, it can back-project 2D cross-sectional PAM and OCT images onto the microscopic view plane. In this way, both microscopic and cross-sectional PAM and OCT images are concurrently displayed on the ocular lens of the microscope. To verify the usability of the NIR-VISPAOCT system, we demonstrate simulated surgeries, including in vivo image-guided melanoma resection surgery and in vivo needle injection of carbon particles into a mouse thigh. The proposed NIR-VISPAOCT system has potential applications in neurosurgery, ophthalmological surgery, and other microsurgeries.


Subject(s)
Microscopy, Acoustic/methods , Photoacoustic Techniques/methods , Surgery, Computer-Assisted/methods , Tomography, Optical Coherence/methods , Animals , Computer Simulation , Drug Delivery Systems/instrumentation , Drug Delivery Systems/methods , Equipment Design , Infrared Rays , Melanoma, Experimental/diagnostic imaging , Melanoma, Experimental/surgery , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Acoustic/instrumentation , Microsurgery/instrumentation , Microsurgery/methods , Photoacoustic Techniques/instrumentation , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/surgery , Surgery, Computer-Assisted/instrumentation , Tomography, Optical Coherence/instrumentation , User-Computer Interface
6.
Stress ; 19(5): 528-34, 2016 09.
Article in English | MEDLINE | ID: mdl-27416924

ABSTRACT

Accumulated evidence indicates that sympathetic nerves may potentiate tumor growth, including melanoma. To elucidate possible mechanisms for this effect, we performed chemical sympathectomy by intraperitoneal (i.p.) injection of the neurotoxin 6-hydroxydopamine hydrobromide (100 mg/kg of body weight); in nine adult male C57BL/6J mice; nine control mice received i.p. vehicle (VEH). Seven days later, all mice were injected subcutaneously with 3 × 10(3) B16-F10 melanoma cells. Mice were euthanized 20 d after injection of melanoma cells, for measurement of tumor weight and expression of genes related to sympathetic signaling, apoptosis, hypoxia and angiogenesis in tumor tissue. To assess potential involvement of the hypothalamo-pituitary-adrenocortical axis in the effect of sympathectomy on melanoma growth, concentrations of plasma corticosterone and level of glucocorticoid receptor mRNA in tumor tissue were determined. We found that sympathectomy significantly attenuated melanoma growth (tumor weight 0.29 ± 0.16 g versus 1.02 ± 0.30 g in controls; p < 0.05). In tumor tissue from sympathectomized mice, we found significantly increased gene expression (measured by real-time PCR), relative to VEH-injected controls, of tyrosine hydroxylase, neuropeptide Y and glucocorticoid receptor (all p < 0.05), and alpha1, beta1 and beta3 adrenergic receptors (all p < 0.025), and factors related to apoptosis (Bcl-2 and caspase-3; p < 0.05) and hypoxia (hypoxia inducible factor 1 alpha) (p = 0.005). Plasma corticosterone concentrations were significantly elevated (p < 0.05) in these mice. Our findings indicate that sympathectomy induces complex changes in the tumor microenvironment reducing melanoma growth. Such complex changes should be considered in the prediction of responses of cancer patients to interventions affecting sympathetic signaling in tumor tissue and its environment.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma, Experimental/surgery , Sympathetic Nervous System/surgery , Animals , Apoptosis/physiology , Caspase 3/metabolism , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neuropeptide Y/metabolism , Oxidopamine/metabolism , Real-Time Polymerase Chain Reaction , Sympathectomy, Chemical , Tumor Burden , Tumor Microenvironment , Tyrosine 3-Monooxygenase/metabolism
7.
Eur J Cancer ; 62: 54-61, 2016 07.
Article in English | MEDLINE | ID: mdl-27208904

ABSTRACT

Cryoablative treatment has been widely used for treating cancer. However, the therapeutic efficacies are still controversial. The molecular mechanisms of the cryo-induced immune responses, particularly underlying the ineffectiveness, remain to be fully elucidated. In this study, we identified a new molecular mechanism involved in the cryo failure. We used cryo-ineffective metastatic tumour models that murine melanoma B16-F10 cells were subcutaneously and intravenously implanted into C57BL/6 mice. When the subcutaneous tumours were treated cryoablation on day 7 after tumour implantation, cells expressing activated leucocyte cell adhesion molecule (ALCAM/CD166) were significantly expanded not only locally in the treated tumours but also systemically in spleen and bone marrow of the mice. The cryo-induced ALCAM(+) cells including CD45(-) mesenchymal stem/stromal cells, CD11b(+)Gr1(+) myeloid-derived suppressor cells, and CD4(+)Foxp3(+) regulatory T cells significantly suppressed interferon γ production and cytotoxicity of tumour-specific CD8(+) T cells via ALCAM expressed in these cells. This suggests that systemic expansion of the ALCAM(+) cells negatively switches host-immune directivity to the tumour-supportive mode. Intratumoural injection with anti-ALCAM blocking monoclonal antibody (mAb) following the cryo treatment systemically induced tumour-specific CD8(+) T cells with higher cytotoxic activities, resulting in suppression of tumour growth and metastasis in the cryo-resistant tumour models. These suggest that expansion of ALCAM(+) cells is a determinant of limiting the cryo efficacy. Further combination with an immune checkpoint inhibitor anti-CTLA4 mAb optimized the anti-tumour efficacy of the dual-combination therapy. Targeting ALCAM may be a promising strategy for overcoming the cryo ineffectiveness leading to the better practical use of cryoablation in clinical treatment of cancer.


Subject(s)
Ablation Techniques/methods , Activated-Leukocyte Cell Adhesion Molecule/physiology , Cryosurgery/methods , Melanoma, Experimental/surgery , Skin Neoplasms/surgery , Tumor Microenvironment/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, CD , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules, Neuronal , Cell Line, Tumor , Fetal Proteins , Flow Cytometry , Interferon-gamma/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Skin Neoplasms/pathology , T-Lymphocytes, Regulatory/immunology
8.
Oncotarget ; 7(14): 18558-72, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26701857

ABSTRACT

Malignant melanoma requires precise resection in order to avoid metastatic recurrence. We report here that the telomerase-dependent, green fluorescent protein (GFP)-containing adenovirus OBP-401 could label malignant melanoma with GFP in situ in orthotopic mouse models. OBP-401-based fluorescence-guided surgery (FGS) resulted in the complete resection of malignant melanoma in the orthotopic models, where conventional bright-light surgery (BLS) could not. High-dose administration of OBP-401 enabled FGS without residual cancer cells or recurrence, due to its dual effect of cancer-cell labeling with GFP and killing.


Subject(s)
Adenoviridae/metabolism , Melanoma, Experimental/surgery , Optical Imaging/methods , Animals , Cell Line, Tumor , Disease Models, Animal , Fluorescence , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Melanoma, Experimental/virology , Mice , Mice, Nude , Neoplasm Recurrence, Local/prevention & control
9.
Anticancer Res ; 35(8): 4425-31, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26168482

ABSTRACT

BACKGROUND/AIM: The outcome of patients with malignant tumors is poor if they suffer from lung metastases. Myeloid-derived suppressor cells (MDSCs), a major player for tumor-induced immunosuppression, can be suppressed by certain chemotherapeutic agents, such as low-dose 5-fluorouracil (5-FU) or surgical treatment. Based on these findings, we hypothesized that early-phase treatment by low-dose 5-FU or surgical resection of primary tumors would prevent lung metastasis formation by inhibiting MDSCs. MATERIALS AND METHODS: B16F10 melanoma-bearing C57BL/5 mice with lung metastases were treated with low-dose 5-FU or surgical resection of primary tumors. RESULTS: Low-dose 5-FU chemotherapy inhibited systemic and lung-accumulating MDSCs in tumor-bearing mice. The therapy inhibited lung metastasis formation and prolonged the survival of the animals. Consistently, early-phase resection of primary tumors improved survival, which was concomitant with a reduction of lung-accumulating MDSCs and lung metastases. CONCLUSION: Early-phase treatment may provide therapeutic values to prevent MDSC-mediated lung metastasis formation in tumor-bearing hosts.


Subject(s)
Antineoplastic Agents/therapeutic use , Fluorouracil/therapeutic use , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma, Experimental/drug therapy , Melanoma, Experimental/surgery , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Female , Fluorouracil/administration & dosage , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/pathology , Secondary Prevention , Treatment Outcome , Tumor Burden/drug effects
10.
Oncotarget ; 6(26): 22857-68, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-25762633

ABSTRACT

Surgical resection of tumors is often followed by regrowth at the primary site and metastases may emerge rapidly following removal of the primary tumor. Macrophages are important drivers of tumor growth, and here we investigated their involvement in postoperative relapse as well as explore macrophage depletion as an adjuvant to surgical resection. RETAAD mice develop spontaneous metastatic melanoma that begins in the eye. Removal of the eyes as early as 1 week of age did not prevent the development of metastases; rather, surgery led to increased proliferation of tumor cells locally and in distant metastases. Surgery-induced increase in tumor cell proliferation correlated with increased macrophage density within the tumor. Moreover, macrophages stimulate tumor sphere formation from tumor cells of post-surgical but not control mice. Macrophage depletion with a diet containing the CSF-1R specific kinase inhibitor Ki20227 following surgery significantly reduced postoperative tumor recurrence and abrogated enhanced metastatic outgrowth. Our results confirm that tumor cells disseminate early, and show that macrophages contribute both to post-surgical tumor relapse and growth of metastases, likely through stimulating a population of tumor-initiating cells. Thus macrophage depletion warrants exploration as an adjuvant to surgical resection.


Subject(s)
Macrophages/pathology , Melanoma, Experimental/pathology , Melanoma, Experimental/surgery , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Macrophages/metabolism , Male , Melanoma, Experimental/metabolism , Mice , Mice, Transgenic , Neoplasm Recurrence, Local/prevention & control , Proto-Oncogene Proteins c-ret/genetics , Surgical Procedures, Operative/methods
11.
Exp Oncol ; 36(1): 24-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24691280

ABSTRACT

UNLABELLED: The aim of the work was experimental study of anticancer efficacy of xenogeneic cancer vaccine (XCV) developed on the basis of rat embryonic nervous tissue and protein-containing metabolite of Bacillus subtilis В-7015 (70 kDa), in В-16 melanoma-bearing С57Bl/6 mice. METHODS: Immunological methods and methods of experimental oncology were used. Effects of XCV on primary and secondary organs of immune system of experimental animals, its anticancer and antimetastatic efficacy were evaluated. RESULTS: It has been shown that XCV did not induced toxic effects on organism, and did not caused inflammatory reactions. The relation between the degree of XCV anticancer efficacy with the regimen of its use and the presence of primary tumor has been analyzed. It has been demonstrated that the developed XCV possesses significant antimetastatic activity if it is used after surgical removal of the primary tumor: in this case lung metastasis inhibition index reached 97.4%. CONCLUSION: High immunogenecity of new XCV creates perspectives for detailed study of its mechanisms of action.


Subject(s)
Cancer Vaccines/administration & dosage , Immunotherapy , Melanoma, Experimental/immunology , Neoplasm Metastasis/immunology , Animals , Antigens, Differentiation/immunology , Bacillus subtilis/immunology , Bacillus subtilis/metabolism , Cancer Vaccines/immunology , Cell Proliferation/drug effects , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/surgery , Mice , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Rats
12.
Melanoma Res ; 24(2): 108-19, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24500501

ABSTRACT

Melanomas are highly lethal skin tumours that are frequently treated by surgical resection. However, the efficacy of such procedures is often limited by tumour recurrence and metastasis. Caveolin-1 (CAV1) has been attributed roles as a tumour suppressor, although in late-stage tumours, its presence is associated with enhanced metastasis. The expression of this protein in human melanoma development and particularly how the presence of CAV1 affects metastasis after surgery has not been defined. CAV1 expression in human melanocytes and melanomas increases with disease progression and is highest in metastatic melanomas. The effect of increased CAV1 expression can then be evaluated using B16F10 murine melanoma cells injected into syngenic immunocompetent C57BL/6 mice or human A375 melanoma cells injected into immunodeficient B6Rag1-/- mice. Augmented CAV1 expression suppresses tumour formation upon a subcutaneous injection, but enhances lung metastasis of cells injected into the tail vein in both models. A procedure was initially developed using B16F10 melanoma cells in C57BL/6 mice to mimic better the situation in patients undergoing surgery. Subcutaneous tumours of a defined size were removed surgically and local tumour recurrence and lung metastasis were evaluated after another 14 days. In this postsurgery setting, CAV1 presence in B16F10 melanomas favoured metastasis to the lung, although tumour suppression at the initial site was still evident. Similar results were obtained when evaluating A375 cells in B6Rag1-/- mice. These results implicate CAV1 expression in melanomas as a marker of poor prognosis for patients undergoing surgery as CAV1 expression promotes experimental lung metastasis in two different preclinical models.


Subject(s)
Caveolin 1/biosynthesis , Melanoma, Experimental/metabolism , Melanoma, Experimental/surgery , Skin Neoplasms/surgery , Animals , Cell Line, Tumor , Humans , Lung Neoplasms/secondary , Melanoma/metabolism , Melanoma/pathology , Melanoma/surgery , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
13.
Clin Cancer Res ; 19(18): 5104-15, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23881927

ABSTRACT

PURPOSE: Surgical removal of solid primary tumors is an essential component of cancer treatment. Surgery-induced dysfunction in natural killer (NK) cells has been linked to the development of metastases in animal models and patients with cancer. We investigated the activation of NK cells using influenza vaccine in the perioperative period to eradicate micrometastatic disease. EXPERIMENTAL DESIGN: Both the B16lacZ and 4T1 tumor models in immunocompetent mice were used to assess the in vivo efficacy of perioperative influenza vaccine administration. In healthy human donors and cancer surgery patients, we assessed NK cell function pre- and post-influenza vaccination using both in vivo and ex vivo assays. RESULTS: Using the TLR3 agonist poly(I:C), we showed as proof-of-principle that perioperative administration of a nonspecific innate immune stimulant can inhibit surgery-induced dysfunction in NK cells and attenuate metastases. Next, we assessed a panel of prophylactic vaccines for NK cell activation and determined that inactivated influenza vaccine was the best candidate for perioperative administration. Perioperative influenza vaccine significantly reduced tumor metastases and improved NK cytotoxicity in preclinical tumor models. Significantly, IFNα is the main cytokine mediator for the therapeutic effect of influenza vaccination. In human studies, influenza vaccine significantly enhanced NK cell activity in healthy human donors and cancer surgery patients. CONCLUSION: These results provide the preclinical rationale to pursue future clinical trials of perioperative NK cell activation, using vaccination in cancer surgery patients. Research into perioperative immune therapy is warranted to prevent immune dysfunction following surgery and eradicate metastatic disease.


Subject(s)
Influenza Vaccines/therapeutic use , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Melanoma, Experimental/surgery , Surgical Procedures, Operative/adverse effects , Animals , Cytotoxicity, Immunologic/immunology , Humans , Lung Neoplasms/secondary , Lung Neoplasms/surgery , Lymphocyte Activation , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Perioperative Care , Postoperative Period , Receptor, Interferon alpha-beta/physiology , Tumor Cells, Cultured , Vaccination
14.
Clin Nutr ; 31(3): 396-404, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22122868

ABSTRACT

BACKGROUND & AIMS: Omega-3 fatty acids (ω-3FA) attenuate postoperative immunosuppression vis-à-vis infection. Since immune-surveillance targets metastasizing cancer cells, we assessed the effect of ω-3FA consumption on 1) early post-operative Natural Killer cell (NK) cytotoxicity and metastases and 2) long-term recurrence-free survival, in two rodent models of surgery-promoted metastases. METHODS: C57BL/6J mice were fed standard, ω-3FA-enriched, or ω-6FA-enriched chow, beginning one week before subcutaneous footpad implantation of syngeneic melanoma cells. When tumors reached the volume of 110 µl, the tumor-bearing footpad was amputated, and long-term recurrence-free survival was assessed. Also, F344 rats were fed ω-3FA or ω-6FA for a month before undergoing or not undergoing laparotomy, and were intravenously inoculated with radio-labeled syngeneic adenocarcinoma cells. Marginating-pulmonary (MP)-leukocytes were harvested, and lung tumor retention (LTR) of metastases was assessed. RESULTS: ω-3FA consumption did not affect the growth of footpad tumors, but significantly enhanced post-amputation recurrence-free survival in mice. Surgery had a deleterious effect on NK cell activity and LTR whereas ω-3FA had large beneficial effects in non-operated rats and an even greater impact in operated rats. CONCLUSIONS: ω-3FA feeding attenuates or even overcomes postoperative NK cell suppression, increases resistance to experimental and spontaneous metastasis, and enhances recurrence-free survival following excision of metastasizing primary tumors. These findings warrant clinical studies of ω-3FA-based nutrition in patients undergoing resection of a primary tumor.


Subject(s)
Fatty Acids, Omega-3/therapeutic use , Fish Oils/therapeutic use , Immunologic Surveillance , Neoplasm Seeding , Neoplasms, Experimental/immunology , Neoplasms, Experimental/prevention & control , Adenocarcinoma/diet therapy , Adenocarcinoma/immunology , Adenocarcinoma/secondary , Adenocarcinoma/surgery , Animals , Combined Modality Therapy , Cytotoxicity, Immunologic , Female , Killer Cells, Natural/immunology , Leukocytes/immunology , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Melanoma, Experimental/secondary , Melanoma, Experimental/surgery , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/diet therapy , Neoplasms, Experimental/surgery , Random Allocation , Rats , Rats, Inbred F344 , Secondary Prevention , Survival Analysis
15.
J Immunol ; 186(6): 3309-16, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21289306

ABSTRACT

A main goal of cancer immunology research is the formation of Ag-specific memory T cell immunity capable of activation upon tumor re-encounter. The requirements necessary to overcome the inhibitory signals present in the tumor microenvironment and form such memory T cell responses are unknown. In contrast to previous studies targeting tumors expressing highly immunogenic model Ags, we demonstrate that alleviating tumor-induced suppression along with vaccination against authentic Ags during the perioperative period provides long-lasting protection against a highly suppressive and poorly immunogenic melanoma. In this study, we employed DNA vaccination with an immunologically optimized mouse melanoma-shared Ag, Trp1ee/ng, combined with systemic TGF-ß blockade during the perioperative period of primary tumor resection, to confer protection against B16 melanoma, and against JBRH, an independently derived melanoma unrelated to B16. Importantly, we demonstrate that correlative to memory responses, perioperative immunotherapy increases the formation of tumor-infiltrating and tumor-reactive CD8(+) T cells expressing low levels of the transcription factor T-bet, defined as memory precursor effector cells. We show that conditions for an immunologically fertile environment are met when TGF-ß blockade and vaccination are applied during the perioperative period of primary tumor resection. These findings address limitations of current CD8(+) T cell immunotherapies against cancer by generating effective CD8(+) T cell memory recall responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Immunologic Memory , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/surgery , Melanoma, Experimental/therapy , Membrane Glycoproteins/therapeutic use , Oxidoreductases/therapeutic use , Transforming Growth Factor beta/antagonists & inhibitors , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/surgery , Drug Therapy, Combination , Immunization, Secondary/methods , Immunologic Memory/genetics , Immunotherapy, Adoptive/methods , Lymphocytes, Tumor-Infiltrating/pathology , Male , Melanoma, Experimental/pathology , Membrane Glycoproteins/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidoreductases/administration & dosage , Perioperative Period/methods , Stem Cells/immunology , Stem Cells/pathology , Transforming Growth Factor beta/physiology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/therapeutic use
16.
J Immunol ; 184(5): 2449-57, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20124103

ABSTRACT

Clinical practice does not consider perioperative paracrine and neuroendocrine stress responses as risk factors for cancer recurrence, although recent animal studies provided supportive evidence. Suggested mechanisms include the effects of stress-hormones on tumor cells and on host physiology. In this study, in mice undergoing primary tumor excision, we tested the survival-enhancing potential of perioperative blockade of catecholamines and prostaglandins, and studied potential mediating mechanisms. C57BL/6J mice were inoculated intrafootpad with syngeneic B16F10.9-melanoma or Lewis lung carcinoma, and the paw was amputated when a developing tumor exceeded 100 microl. The clinically used beta-adrenergic antagonist propranolol, and/or the cyclooxygenase-2 inhibitor etodolac, were administered once before amputation, and recurrence-free survival was monitored. In different studies, NK cytotoxicity, leukocytes' molecular functional markers, and vascular endothelial growth factor secretion by tumor cells were studied in the context of surgery and drug treatments. The findings indicated that the combination of propranolol and etodolac, but neither drug alone, significantly and markedly improved survival rates in both tumor models, and was as effective as established immunostimulatory agents (IL-12 and polyinosinic-polycytiylic acid). Surgery markedly reduced NK cytotoxicity and NK cell expression of Fas ligand and CD11a, reduced all circulating lymphocyte-subtype concentrations, and increased corticosterone levels. Propranolol and etodolac administration counteracted these perturbations. B16 and 3LL secreted vascular endothelial growth factor in vitro, but secretion was not affected by catecholamine agonists, prostaglandins, corticosterone, propranolol, or etodolac. Overall, propranolol and etodolac administration, which could be applied perioperatively in most cancer patients with minimal risk and low cost, has counteracted several immunologic and endocrinologic perturbations and improved recurrence-free survival rates in mice undergoing primary tumor excision.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Lewis Lung/surgery , Melanoma, Experimental/surgery , Postoperative Complications/prevention & control , Adrenergic beta-Antagonists/administration & dosage , Amputation, Surgical/adverse effects , Animals , CD11a Antigen/metabolism , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Cyclooxygenase 2 Inhibitors/administration & dosage , Etodolac/administration & dosage , Fas Ligand Protein/metabolism , Female , Interleukin-12/administration & dosage , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Laparotomy/adverse effects , Male , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Poly I-C/administration & dosage , Postoperative Complications/etiology , Postoperative Complications/mortality , Propranolol/administration & dosage , Survival Rate
17.
Cancer Res ; 69(20): 7926-34, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19826056

ABSTRACT

The circulating tumor cell (CTC) count has been shown as a prognostic marker for metastasis development. However, its clinical utility for metastasis prevention remains unclear, because metastases may already be present at the time of initial diagnosis with existing assays. Their sensitivity ex vivo is limited by a small blood sample volume, whereas in vivo examination of larger blood volumes may be clinically restricted by the toxicity of labels used for targeting of CTCs. We introduce a method for in vivo photoacoustic blood cancer testing with a high-pulse-repetition-rate diode laser that, when applied to melanoma, is free of this limitation. It uses the overexpression of melanin clusters as intrinsic, spectrally-specific cancer markers and signal amplifiers, thus providing higher photoacoustic contrast of melanoma cells compared with a blood background. In tumor-bearing mouse models and melanoma-spiked human blood samples, we showed a sensitivity level of 1 CTC/mL with the potential to improve this sensitivity 10(3)-fold in humans in vivo, which is impossible with existing assays. Additional advances of this platform include decreased background signals from blood through changes in its oxygenation, osmolarity, and hematocrit within physiologic norms, assessment of CTCs in deep vessels, in vivo CTC enrichment, and photoacoustic-guided photothermal ablation of CTCs in the bloodstream. These advances make feasible the early diagnosis of melanoma during the initial parallel progression of primary tumor and CTCs, and laser blood purging using noninvasive or hemodialysis-like schematics for the prevention of metastasis.


Subject(s)
Flow Cytometry , Laser Therapy/methods , Lasers, Semiconductor/therapeutic use , Melanins/metabolism , Melanoma, Experimental/surgery , Neoplastic Cells, Circulating/pathology , Skin Neoplasms/surgery , Animals , Humans , Mice , Mice, Transgenic , Neoplastic Cells, Circulating/metabolism , Tumor Cells, Cultured
18.
Cancer Res ; 69(9): 3979-85, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19383917

ABSTRACT

Tumors that recur following surgical resection of melanoma are typically metastatic and associated with poor prognosis. Using the murine B16F10 melanoma and a robust antimelanoma vaccine, we evaluated immunization as a tool to improve tumor-free survival following surgery. We investigated the utility of vaccination in both neoadjuvant and adjuvant settings. Surprisingly, neoadjuvant vaccination was far superior and provided approximately 100% protection against tumor relapse. Neoadjuvant vaccination was associated with enhanced frequencies of tumor-specific T cells within the tumor and the tumor-draining lymph nodes following resection. We also observed increased infiltration of antigen-specific T cells into the area of surgery. This method should be amenable to any vaccine platform and can be readily extended to the clinic.


Subject(s)
Cancer Vaccines/therapeutic use , Melanoma, Experimental/therapy , Neoplasm Recurrence, Local/prevention & control , Adenoviridae/genetics , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Female , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/surgery , Mice , Mice, Inbred C57BL , Neoadjuvant Therapy , Neoplasm Recurrence, Local/immunology , T-Lymphocytes/immunology
19.
Clin Exp Metastasis ; 25(5): 531-6, 2008.
Article in English | MEDLINE | ID: mdl-18357506

ABSTRACT

Although surgical removal is the most aggressive strategy to treat removable tumors, it sometimes aggravates tumor growth in metastatic sites. Because surgical procedures generate reactive oxygen species (ROS), known promoters of tumor metastasis and growth, we examined whether the growth of micrometastasis is inhibited by superoxide dismutase (SOD) derivatives after surgical removal of tumors in mice. Murine melanoma B16-BL6 cells were inoculated into the footpad to establish spontaneous pulmonary metastasis. The removal of the footpad tumor significantly (P < 0.05) increased the level of plasma lipoperoxides and the number of tumor cells in the lung. An intravenous injection of SOD or its pegylated-SOD derivative significantly (P < 0.05) inhibited the peroxidation and metastatic tumor growth. It also extended the survival period of mice undergoing removal of the footpad tumor. These findings indicate that the removal of tumor produces ROS, which then aggravates the growth of tumor cells in micrometastases. SOD derivatives can effectively prevent this metastatic tumor growth by detoxifying ROS.


Subject(s)
Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma, Experimental/secondary , Melanoma, Experimental/surgery , Superoxide Dismutase/therapeutic use , Animals , Lipid Peroxides/blood , Lung Neoplasms/blood , Male , Melanoma, Experimental/blood , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Reactive Oxygen Species/metabolism
20.
Cancer Res ; 68(6): 1614-7, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18339838

ABSTRACT

Antitumor immune responses can be stimulated by interfering with regulatory T-cell (T(reg)) function. However, this effect is short lived unless T-cell memory to tumor antigens can be generated. Our recent studies show that T(reg) cells not only limit primary responses to tumor/self-antigens in tumor-bearing hosts but also prevent the natural generation of T-cell memory to such antigens. Here, we discuss the role of T(reg) cells in suppressing T-cell memory after surgical excision of tumors and the potential clinical benefits of overcoming this suppression.


Subject(s)
Immunologic Memory , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Neoplasm/immunology , Humans , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/surgery , Mice , Neoplasms/pathology , Neoplasms/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...