Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cells ; 12(20)2023 10 11.
Article in English | MEDLINE | ID: mdl-37887283

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors have transformed clinical oncology. However, their use is limited as response is observed in only ~20-50% of patients. Previously, we demonstrated that treating CT26 tumor-bearing mice with ultra-high-concentration gaseous nitric oxide (UNO) followed by tumor resection stimulated antitumor immune responses. Accordingly, UNO may improve tumor response to immune checkpoint inhibitors. Here, we investigated the ability of UNO to improve the efficacy of a programmed cell death protein-1 (PD-1) antibody in vitro and in treating CT26 tumor-bearing mice. METHODS: CT26 cells were injected into the flank of Balb/c mice (n = 15-16 per group). On day 6, CT26 cells were injected into the contralateral flank, and anti-mPD-1 injections commenced. Primary tumors were treated with intratumoral UNO on day 8. Tumor volume, response rates, toxicity, and survival were monitored. RESULTS: (1) Short exposure to 25,000-100,000 parts per million (ppm) UNO in vitro resulted in significant upregulation of PD-L1 expression on CT26 cells. (2) UNO treatment in vivo consistently reduced cell viability in CT26 tumors. (3) Treatment reduced regulatory T-cell (Treg) levels in the tumor and increased levels of systemic M1 macrophages. UNO responders had increased CD8+ T-cell tumor infiltration. (4) Nine days after treatment, primary tumor growth was significantly lower in the combination arm vs. anti-mPD-1 alone (p = 0.0005). (5) Complete tumor regression occurred in 8/15 (53%) of mice treated with a combination of 10 min UNO and anti-mPD-1, 100 days post-treatment, compared to 4/16 (25%) of controls treated with anti-mPD-1 alone (p = 0.1489). (6) There was no toxicity associated with UNO treatment. (7) Combination treatment showed a trend toward increased survival 100 days post-treatment compared to anti-mPD-1 alone (p = 0.0653). CONCLUSION: Combining high-concentration NO and immune checkpoint inhibitors warrants further assessment especially in tumors resistant to checkpoint inhibitor therapy.


Subject(s)
Immune Checkpoint Inhibitors , Nitric Oxide , Humans , Mice , Animals , Nitric Oxide/metabolism , Cell Line, Tumor , CD8-Positive T-Lymphocytes
2.
Cancer Cell Int ; 22(1): 405, 2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36514083

ABSTRACT

BACKGROUND: In-situ tumor ablation provides the immune system with the appropriate antigens to induce anti-tumor immunity. Here, we present an innovative technique for generating anti-tumor immunity by delivering exogenous ultra-high concentration (> 10,000 ppm) gaseous nitric oxide (UHCgNO) intratumorally. METHODS: The capability of UHCgNO to induce apoptosis was tested in vitro in mouse colon (CT26), breast (4T1) and Lewis lung carcinoma (LLC-1) cancer cell lines. In vivo, UHCgNO was studied by treating CT26 tumor-bearing mice in-situ and assessing the immune response using a Challenge assay. RESULTS: Exposing CT26, 4T1 and LLC-1 cell lines to UHCgNO for 10 s-2.5 min induced cellular apoptosis 24 h after exposure. Treating CT26 tumors in-situ with UHCgNO followed by surgical resection 14 days later resulted in a significant secondary anti-tumor effect in vivo. 100% of tumor-bearing mice treated with 50,000 ppm UHCgNO and 64% of mice treated with 20,000 ppm UHCgNO rejected a second tumor inoculation, compared to 0% in the naive control for 70 days. Additionally, more dendrocytes infiltrated the tumor 14 days post UHCgNO treatment versus the nitrogen control. Moreover, T-cell penetration into the primary tumor was observed in a dose-dependent manner. Systemic increases in T- and B-cells were seen in UHCgNO-treated mice compared to nitrogen control. Furthermore, polymorphonuclear-myeloid-derived suppressor cells were downregulated in the spleen in the UHCgNO-treated groups. CONCLUSIONS: Taken together, our data demonstrate that UHCgNO followed by the surgical removal of the primary tumor 14 days later induces a strong and potent anti-tumor response.

3.
Cancer Immunol Immunother ; 65(10): 1149-58, 2016 10.
Article in English | MEDLINE | ID: mdl-27495172

ABSTRACT

It has been demonstrated that aggressive in situ tumor destruction (ablation) could lead to the release of tumor antigens, which can stimulate anti-tumor immune responses. We developed an innovative method of tumor ablation based on intratumoral alpha-irradiation, diffusing alpha-emitters radiation therapy (DaRT), which efficiently ablates local tumors and enhances anti-tumor immunity. In this study, we investigated the anti-tumor potency of a treatment strategy, which combines DaRT tumor ablation with two approaches for the enhancement of anti-tumor reactivity: (1) neutralization of immunosuppressive cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) and (2) boost the immune response by the immunoadjuvant CpG. Mice bearing DA3 mammary adenocarcinoma with metastases were treated with DaRT wires in combination with a MDSC inhibitor (sildenafil), Treg inhibitor (cyclophosphamide at low dose), and the immunostimulant, CpG. Combination of all four therapies led to a complete rejection of primary tumors (in 3 out of 20 tumor-bearing mice) and to the elimination of lung metastases. The treatment with DaRT and Treg or MDSC inhibitors (without CpG) also resulted in a significant reduction in tumor size, reduced the lung metastatic burden, and extended survival compared to the corresponding controls. We suggest that the therapy with DaRT combined with the inhibition of immunosuppressive cells and CpG reinforced both local and systemic anti-tumor immune responses and displayed a significant anti-tumor effect in tumor-bearing mice.


Subject(s)
Adenocarcinoma/therapy , Alpha Particles/therapeutic use , Brachytherapy , Breast Neoplasms/therapy , Cell Growth Processes , Cyclophosphamide/therapeutic use , Myeloid-Derived Suppressor Cells/immunology , Sildenafil Citrate/therapeutic use , T-Lymphocytes, Regulatory/immunology , Ablation Techniques/statistics & numerical data , Animals , Antigens, Neoplasm/immunology , Cell Growth Processes/drug effects , Cell Growth Processes/radiation effects , Cell Line, Tumor , Combined Modality Therapy , Female , Humans , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/therapeutic use , Tumor Burden/drug effects , Tumor Burden/radiation effects
4.
Int J Radiat Biol ; 91(2): 179-86, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25179346

ABSTRACT

PURPOSE: We developed (224)Ra-loaded wires, which release by recoil alpha emitting nuclei into solid tumors and cause tumor cell killing. This research examined if the major damage was inflicted by alpha particles emitted from these atoms or by direct gamma and beta emissions from the inserted wires. We also examined the efficacy of this treatment against colon cancer in combination with chemotherapy. MATERIALS AND METHODS: Mouse colon carcinomas (CT-26 xenografts), treated by intra-tumoral radioactive wires loaded with (224)Ra atoms were monitored for effects on tumor growth, intratumoral tissue damage and distribution of alpha emitting atoms. The effects were compared with those of (224)Ra-loaded wires coated with poly methyl methacrylate (PMMA), which blocks atom recoil. Similar experiments were performed with radioactive wires combined with systemic 5-FU. RESULTS: (224)Ra-loaded wires inhibited tumor growth and formed necrotic areas inside the tumor. PMMA coated wires did not inhibit tumor growth, and caused minor intratumoral damage. Autoradiography images of tumors treated with (224)Ra-loaded wires revealed a spread of alpha emitters over several mm, whereas PMMA-coated wires showed no such spread. Injection of 5-FU with (224)Ra-loaded wires augmented tumor growth retardation and cure. CONCLUSIONS: (224)Ra-loaded wires ablate solid tumors by the release of alpha-particle emitting atoms inside the tissue, an effect that can be enhanced by combining this method with chemotherapy.


Subject(s)
Ablation Techniques/instrumentation , Alpha Particles/therapeutic use , Antineoplastic Agents/pharmacology , Brachytherapy/instrumentation , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Radium/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/surgery , Combined Modality Therapy , Disease Models, Animal , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Humans , Male , Mice
5.
Cancer Immunol Immunother ; 64(2): 191-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25326364

ABSTRACT

INTRODUCTION: The current systemic anti-metastatic treatment is chemotherapy. Chemotherapy reacts mostly against replicating cells, which makes this therapy not specific. Moreover, resting cancer cells will not be destroyed. A better alternative is an engagement of the host immune system to react against tumor-associated antigens. An efficient immune-stimulating technique is an ablation of the tumor that results in the release of tumor antigens. Our ablation strategy is an innovative alpha-radiation-based technology, diffusing alpha-emitters radiation therapy (DaRT), which efficiently destroys local tumors and provides thereby an antigenic supply for antigen-presenting cells to stimulate T cells. METHODS: Mice bearing weakly immunogenic DA3 adenocarcinoma or highly immunogenic CT26 colon carcinoma were treated by DaRT. Anti-tumor immune responses following tumor destruction were evaluated by (1) the resistance to a tumor challenge; (2) scanning by a CT imaging device for elimination of lung metastases; (3) improved tumor control when combining DaRT with an immunoadjuvant (CpG). RESULTS: CT26 model: 63-77 % of DaRT-treated mice became resistant to a re-inoculated tumor compared to 29-33 % resistant mice in the control. DA3 model: (1) The growth rate of challenge tumors was the lowest in mice which their primary tumor was treated by DaRT. (2) Most (93 %) mice in the control group developed lung metastases compared to 56 % in the DaRT group. (3) Combining DaRT with CpG resulted in a better control of the primary tumor. Our study offers a technique to eliminate local and distant malignant cells, regardless of their replication status, by stimulating specific anti-tumor immunity through the supply of tumor antigens from the destroyed tumor.


Subject(s)
Ablation Techniques , Alpha Particles/therapeutic use , Neoplasms, Experimental/immunology , Neoplasms, Experimental/radiotherapy , Adjuvants, Immunologic/therapeutic use , Animals , Antigens, Neoplasm/immunology , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Female , Humans , Lung Neoplasms/secondary , Male , Mice , Neoplasm Metastasis , Neoplasms, Experimental/pathology , Oligodeoxyribonucleotides/administration & dosage , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
6.
Cancer Immunol Immunother ; 63(1): 1-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23955682

ABSTRACT

Cancer, the most devastating chronic disease affecting humankind, is treated primarily by surgery, chemotherapy, and radiation therapy. Surgery and radiotherapy are mainly used for debulking the primary tumor, while chemotherapy is the most efficient anti-metastatic treatment. To control better metastatic cancer, the host immune system should be stimulated. Yet, successful specific stimulation of the immune system against tumors was seldom achieved even in antigenic tumors. Our working hypothesis is that aggressive in situ tumor ablation can release tumor antigens and danger signals, which will enhance anti-tumor T cell responses resulting in the destruction of residual malignant cells in primary tumors and distant metastases. We developed two efficient in situ ablation treatments for solid cancer, which can be used to destroy the primary tumors and stimulate anti-tumor immune responses. The first treatment, electrochemical ablation, is applied through intratumoral electrodes, which deliver unipolar-pulsed electric currents. The second treatment, diffusing alpha-emitters radiation therapy (DaRT), is based on intratumoral (224)Ra-loaded wire(s) that release by recoil its daughter atoms. These short-lived alpha-emitting atoms spread in the tumor and spray it with lethal alpha particles. It was confirmed that these treatments effectively destroy various malignant animal and human primary solid tumors. As a consequence of such tumor ablation, tumor-derived antigenic material was released and provoked systemic T cell-dependent anti-tumor immunological reactions. These reactions conferred protection against a secondary tumor challenge and destroyed remaining malignant cells in the primary tumor as well as in distant metastases. Such anti-tumor immune responses could be further amplified by the immune adjuvant, CpG. Electrochemical ablation or DaRT together with chemotherapy and immunostimulatory agents can serve as treatment protocols for solid metastatic tumors and can be applied instead of or in combination with surgery.


Subject(s)
Ablation Techniques/methods , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/radiotherapy , Alpha Particles , Animals , Antigens, Neoplasm/chemistry , Antineoplastic Agents/chemistry , Electrochemistry , Humans , Immunologic Memory , Mice , Neoplasm Metastasis , Radium/chemistry , Time Factors
7.
J Biomed Opt ; 18(11): 111410, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23934014

ABSTRACT

Treating cancer is one of the major challenges of modern medicine. Since mice models are an important tool in cancer treatment research, it is required to assess murine tumor development. Existing methods for investigating tumor development are either high cost and limited by their availability or suffer from low accuracy and reproducibility. In order to overcome these drawbacks, thermography may be used. DA3 breast cancer carcinoma tumors in 12 Balb/c mice were thermally imaged and monitored for a period of several weeks. Eight mice were treated with diffusing alpha emitters radiation therapy (DaRT) wires, while four were treated with inert wires. For large tumors, the area was estimated by analyzing thermal images and was found to be in correlation with manual caliper measurements. In addition, the correlation between tumor area and relative temperatures was calculated and compared to previous works. Temperature differences were larger for tumors treated with DaRT wires than tumors with inert wires. These correlations can be used to assist in tumor size estimation and reveal information regarding its metabolic state. Overall, thermography was shown to be a promising tool for assessing tumor development with the additional advantages of being nonradiative and potentially providing indication of intratumoral biological processes.


Subject(s)
Body Temperature/physiology , Mammary Neoplasms, Experimental/physiopathology , Thermography/methods , Algorithms , Animals , Female , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/radiotherapy , Mice , Mice, Inbred BALB C , Necrosis
SELECTION OF CITATIONS
SEARCH DETAIL
...