1
|
Mabrouk N, Racoeur C, Shan J, Massot A, Ghione S, Privat M, Dondaine L, Ballot E, Truntzer C, Boidot R, Hermetet F, Derangère V, Bruchard M, Végran F, Chouchane L, Ghiringhelli F, Bettaieb A, Paul C. GTN Enhances Antitumor Effects of Doxorubicin in TNBC by Targeting the Immunosuppressive Activity of PMN-MDSC. Cancers (Basel) 2023; 15:3129. [PMID: 37370739 DOI: 10.3390/cancers15123129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/17/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
(1) Background: Immunosuppression is a key barrier to effective anti-cancer therapies, particularly in triple-negative breast cancer (TNBC), an aggressive and difficult to treat form of breast cancer. We investigated here whether the combination of doxorubicin, a standard chemotherapy in TNBC with glyceryltrinitrate (GTN), a nitric oxide (NO) donor, could overcome chemotherapy resistance and highlight the mechanisms involved in a mouse model of TNBC. (2) Methods: Balb/C-bearing subcutaneous 4T1 (TNBC) tumors were treated with doxorubicin (8 mg/Kg) and GTN (5 mg/kg) and monitored for tumor growth and tumor-infiltrating immune cells. The effect of treatments on MDSCs reprogramming was investigated ex vivo and in vitro. (3) Results: GTN improved the anti-tumor efficacy of doxorubicin in TNBC tumors. This combination increases the intra-tumor recruitment and activation of CD8+ lymphocytes and dampens the immunosuppressive function of PMN-MDSCs PD-L1low. Mechanistically, in PMN-MDSC, the doxorubicin/GTN combination reduced STAT5 phosphorylation, while GTN +/- doxorubicin induced a ROS-dependent cleavage of STAT5 associated with a decrease in FATP2. (4) Conclusion: We have identified a new combination enhancing the immune-mediated anticancer therapy in a TNBC mouse model through the reprograming of PMN-MDSCs towards a less immunosuppressive phenotype. These findings prompt the testing of GTN combined with chemotherapies as an adjuvant in TNBC patients experiencing treatment failure.
Collapse
Affiliation(s)
- Nesrine Mabrouk
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75006 Paris, France
- LIIC, EA7269, Université de Bourgogne Franche Comté, 21000 Dijon, France
| | - Cindy Racoeur
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75006 Paris, France
- LIIC, EA7269, Université de Bourgogne Franche Comté, 21000 Dijon, France
| | - Jingxuan Shan
- Genetic Intelligence Laboratory, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha P.O. Box 24144, Qatar
| | - Aurélie Massot
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75006 Paris, France
- LIIC, EA7269, Université de Bourgogne Franche Comté, 21000 Dijon, France
| | - Silvia Ghione
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75006 Paris, France
- LIIC, EA7269, Université de Bourgogne Franche Comté, 21000 Dijon, France
| | - Malorie Privat
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75006 Paris, France
- LIIC, EA7269, Université de Bourgogne Franche Comté, 21000 Dijon, France
| | - Lucile Dondaine
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75006 Paris, France
- LIIC, EA7269, Université de Bourgogne Franche Comté, 21000 Dijon, France
| | - Elise Ballot
- Plateforme de Transfert en Biologie Cancérologique, Centre GFL Leclerc, 21000 Dijon, France
| | - Caroline Truntzer
- Plateforme de Transfert en Biologie Cancérologique, Centre GFL Leclerc, 21000 Dijon, France
| | - Romain Boidot
- Unit of Molecular Biology, Georges-François Leclerc Cancer Center-UNICANCER, CNRS UMR 6302, 21000 Dijon, France
| | | | - Valentin Derangère
- Plateforme de Transfert en Biologie Cancérologique, Centre GFL Leclerc, 21000 Dijon, France
- UBFC, 21000 Dijon, France
| | - Mélanie Bruchard
- CRI UMR INSERM1231, 21000 Dijon, France
- UBFC, 21000 Dijon, France
| | - Frédérique Végran
- Plateforme de Transfert en Biologie Cancérologique, Centre GFL Leclerc, 21000 Dijon, France
- CRI UMR INSERM1231, 21000 Dijon, France
- UBFC, 21000 Dijon, France
| | - Lotfi Chouchane
- Genetic Intelligence Laboratory, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha P.O. Box 24144, Qatar
| | - François Ghiringhelli
- Plateforme de Transfert en Biologie Cancérologique, Centre GFL Leclerc, 21000 Dijon, France
- CRI UMR INSERM1231, 21000 Dijon, France
- UBFC, 21000 Dijon, France
| | - Ali Bettaieb
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75006 Paris, France
- LIIC, EA7269, Université de Bourgogne Franche Comté, 21000 Dijon, France
| | - Catherine Paul
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75006 Paris, France
- LIIC, EA7269, Université de Bourgogne Franche Comté, 21000 Dijon, France
| |
Collapse
|
2
|
Bonavida B. Historical Perspectives of the Role of NO/NO Donors in Anti-Tumor Activities: Acknowledging Dr. Keefer's Pioneering Research. Crit Rev Oncog 2023; 28:1-13. [PMID: 37824383 DOI: 10.1615/critrevoncog.2021035853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The role of nitric oxide (NO) in cancer has been a continuous challenge and particularly the contradictory findings in the literature reporting NO with either anti-cancer properties or pro-cancer properties. This dilemma was largely resolved by the level of NO/inducible nitric oxide synthase in the tumor environment as well as other cancer-associated gene activations in different cancers. The initial findings on the role of NO as an anti-cancer agent was initiated in the late 1990's in Dr. Larry Keefer's laboratory, who had been studying and synthesizing many compounds with releasing NO under different conditions. Using an experimental model with selected NO compounds they demonstrated for the first time that NO can inhibit tumor cell proliferation and sensitizes drug-resistant cancer cells to chemotherapy-induced cytotoxicity. This initial finding was the backbone and the foundation of subsequent reports by the Keefer's laboratory and followed by many others to date on NO-mediated anti-cancer activities and the clinical translation of NO donors in cancer therapy. Our laboratory initiated studies on NO-mediated anti-cancer therapy and chemo-immuno-sensitization following Keefer's findings and used one of his synthesized NO donors, namely, (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate (DETANONOate), throughout most of our studies. Many of Keefer's collaborators and other investigators have reported on the selected compound, O2-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl)piperazin-1-yl] diazen-1-ium-1,2-diolate (JS-K), and its therapeutic role in many tumor model systems. Several lines of evidence that investigated the treatment with NO donors in various cancer models revealed that a large number of gene products are modulated by NO, thus emphasizing the pleiotropic effects of NO on cancers and the identification of many targets of therapeutic significance. The present review reports historically of several examples reported in the literature that emanated on NO-mediated anti-cancer activities by the Keefer's laboratory and his collaborators and other investigators including my laboratory at the University of California at Los Angeles.
Collapse
Affiliation(s)
- Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
3
|
Burke AJ, McAuliffe JD, Natoni A, Ridge S, Sullivan FJ, Glynn SA. Chronic nitric oxide exposure induces prostate cell carcinogenesis, involving genetic instability and a pro-tumorigenic secretory phenotype. Nitric Oxide 2022; 127:44-53. [PMID: 35872082 DOI: 10.1016/j.niox.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 11/26/2022]
Abstract
Prostate cancer is a leading cause of cancer death in men. Inflammation and overexpression of inducible nitric oxide synthase (NOS2) have been implicated in prostate carcinogenesis. We aimed to explore the hypothesis that nitric oxide NO exerts pro-tumorigenic effects on prostate cells at physiologically relevant levels contributing to carcinogenesis. We investigated the impact of acute exposure of normal immortalised prostate cells (RWPE-1) to NO on cell proliferation and activation of DNA damage repair pathways. Furthermore we investigated the long term effects of chronic NO exposure on RWPE-1 cell migration and invasion potential and hallmarks of transformation. Our results demonstrate that NO induces DNA damage as indicated by γH2AX foci and p53 activation resulting in a G1/S phase block and activation of 53BP1 DNA damage repair protein. Long term adaption to NO results in increased migration and invasion potential, acquisition of anchorage independent growth and increased resistance to chemotherapy. This was recapitulated in PC3 and DU145 prostate cancer cells which upon chronic exposure to NO displayed increased cell migration, colony formation and increased resistance to chemotherapeutics. These findings indicate that NO may play a key role in the development of prostate cancer and the acquisition of an aggressive metastatic phenotype.
Collapse
Affiliation(s)
- Amy J Burke
- Prostate Cancer Institute, School of Medicine, National University of Ireland Galway, Galway, H91 TK33, Ireland
| | - Jake D McAuliffe
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, Galway, H91 TK33, Ireland
| | - Alessandro Natoni
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Sarah Ridge
- Prostate Cancer Institute, School of Medicine, National University of Ireland Galway, Galway, H91 TK33, Ireland
| | - Francis J Sullivan
- Prostate Cancer Institute, School of Medicine, National University of Ireland Galway, Galway, H91 TK33, Ireland
| | - Sharon A Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, Galway, H91 TK33, Ireland.
| |
Collapse
|
4
|
Salihi A, Al-Naqshabandi MA, Khudhur ZO, Housein Z, Hama HA, Abdullah RM, Hussen BM, Alkasalias T. Gasotransmitters in the tumor microenvironment: Impacts on cancer chemotherapy (Review). Mol Med Rep 2022; 26:233. [PMID: 35616143 PMCID: PMC9178674 DOI: 10.3892/mmr.2022.12749] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/07/2022] [Indexed: 11/23/2022] Open
Abstract
Nitric oxide, carbon monoxide and hydrogen sulfide are three endogenous gasotransmitters that serve a role in regulating normal and pathological cellular activities. They can stimulate or inhibit cancer cell proliferation and invasion, as well as interfere with cancer cell responses to drug treatments. Understanding the molecular pathways governing the interactions between these gases and the tumor microenvironment can be utilized for the identification of a novel technique to disrupt cancer cell interactions and may contribute to the conception of effective and safe cancer therapy strategies. The present review discusses the effects of these gases in modulating the action of chemotherapies, as well as prospective pharmacological and therapeutic interfering approaches. A deeper knowledge of the mechanisms that underpin the cellular and pharmacological effects, as well as interactions, of each of the three gases could pave the way for therapeutic treatments and translational research.
Collapse
Affiliation(s)
- Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region 44001, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan Region 44002, Iraq
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, SE-17165 Stockholm, Sweden
| | - Mohammed A. Al-Naqshabandi
- Department of Clinical Biochemistry, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region 44001, Iraq
| | - Zhikal Omar Khudhur
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Kurdistan Region 44001, Iraq
| | - Zjwan Housein
- Department of Medical Laboratory Technology, Technical Health and Medical College, Erbil Polytechnique University, Erbil, Kurdistan Region 44002, Iraq
| | - Harmand A. Hama
- Department of Biology, Faculty of Education, Tishk International University, Erbil, Kurdistan Region 44002, Iraq
| | - Ramyar M. Abdullah
- College of Medicine, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Twana Alkasalias
- General Directorate of Scientific Research Center, Salahaddin University-Erbil, Erbil, Kurdistan Region 44002, Iraq
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| |
Collapse
|
5
|
Marullo R, Castro M, Yomtoubian S, Calvo-Vidal MN, Revuelta MV, Krumsiek J, Cho A, Morgado PC, Yang S, Medina V, Roth BM, Bonomi M, Keshari KR, Mittal V, Navigante A, Cerchietti L. The metabolic adaptation evoked by arginine enhances the effect of radiation in brain metastases. SCIENCE ADVANCES 2021; 7:eabg1964. [PMID: 34739311 PMCID: PMC8570607 DOI: 10.1126/sciadv.abg1964] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Selected patients with brain metastases (BM) are candidates for radiotherapy. A lactatogenic metabolism, common in BM, has been associated with radioresistance. We demonstrated that BM express nitric oxide (NO) synthase 2 and that administration of its substrate l-arginine decreases tumor lactate in BM patients. In a placebo-controlled trial, we showed that administration of l-arginine before each fraction enhanced the effect of radiation, improving the control of BM. Studies in preclinical models demonstrated that l-arginine radiosensitization is a NO-mediated mechanism secondary to the metabolic adaptation induced in cancer cells. We showed that the decrease in tumor lactate was a consequence of reduced glycolysis that also impacted ATP and NAD+ levels. These effects were associated with NO-dependent inhibition of GAPDH and hyperactivation of PARP upon nitrosative DNA damage. These metabolic changes ultimately impaired the repair of DNA damage induced by radiation in cancer cells while greatly sparing tumor-infiltrating lymphocytes.
Collapse
Affiliation(s)
- Rossella Marullo
- Hematology and Oncology Division, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Monica Castro
- Translational Research Unit, Angel Roffo Cancer Institute, University of Buenos Aires, Buenos Aires, Argentina
| | - Shira Yomtoubian
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - M. Nieves Calvo-Vidal
- Hematology and Oncology Division, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Maria Victoria Revuelta
- Hematology and Oncology Division, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jan Krumsiek
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Andrew Cho
- Department of Biochemistry and Structural Biology, Weill Cornell Graduate School, New York, NY, USA
| | - Pablo Cresta Morgado
- Translational Research Unit, Angel Roffo Cancer Institute, University of Buenos Aires, Buenos Aires, Argentina
| | - ShaoNing Yang
- Hematology and Oncology Division, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Vanina Medina
- Laboratory of Tumor Biology and Inflammation, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina and National Scientific and Technical Research Council, Buenos Aires, Argentina
- Laboratory of Radioisotopes, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Berta M. Roth
- Radiation and Imaging Department, Angel Roffo Cancer Institute, University of Buenos Aires, Buenos Aires, Argentina
| | - Marcelo Bonomi
- Hematology and Oncology Division, The Ohio State University, Columbus, OH, USA
| | - Kayvan R. Keshari
- Department of Biochemistry and Structural Biology, Weill Cornell Graduate School, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vivek Mittal
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Alfredo Navigante
- Translational Research Unit, Angel Roffo Cancer Institute, University of Buenos Aires, Buenos Aires, Argentina
| | - Leandro Cerchietti
- Hematology and Oncology Division, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Corresponding author.
| |
Collapse
|
6
|
Feng H, Kishimura A, Mori T, Katayama Y. Evaluation of a Synergistic Effect of L-Arginine on the Anticancer Activity of Doxorubicin by Using a Co-culture System. ANAL SCI 2020; 36:1279-1283. [PMID: 32624523 DOI: 10.2116/analsci.20p200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In the early stage of tumor development, tumor-associated macrophages (TAM) works to suppress tumor growth by secreting soluble factors including nitric oxide (NO). L-Arginine (Arg) is a substrate of nitric oxide synthase (NOS) expressed in TAM. Here we examined whether NO produced from Arg by macrophages works to enhance the effect of the anti-cancer drug, doxorubicin (Dox) by using a co-culture system of cancer cells with macrophages. By employing colorimetric analyses methods (Griess Reagent and Cell Counting kit-8), we found that NO produced from Arg by co-cultured macrophages could enhance the cytotoxic effect of Dox to cancer cells. Moreover, we found that augmentation is affected by the order of the addition of Arg and Dox. A prior addition of Arg to Dox and simultaneous addition showed the same enhancement effect, but a prior addition of Dox to Arg abolished the augmentation. This suggests that the co-administration of Arg with Dox would be an effective treatment to improve chemo-therapies.
Collapse
Affiliation(s)
- Haitao Feng
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University
| | - Akihiro Kishimura
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University.,Graduate School of System Life Science, Kyushu University.,International Research Center for Molecular Systems, Kyushu University
| | - Takeshi Mori
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University.,Graduate School of System Life Science, Kyushu University
| | - Yoshiki Katayama
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University.,Graduate School of System Life Science, Kyushu University.,International Research Center for Molecular Systems, Kyushu University.,Center for Advanced Medical Innovation, Kyushu University.,Department of Biomedical Engineering, Chung Yuan Christian University
| |
Collapse
|
7
|
Bonavida B. Sensitizing activities of nitric oxide donors for cancer resistance to anticancer therapeutic drugs. Biochem Pharmacol 2020; 176:113913. [PMID: 32173364 DOI: 10.1016/j.bcp.2020.113913] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/10/2020] [Indexed: 02/08/2023]
Abstract
Cancer is not a single disease but it constitutes a large variety of different types that are also different from each other phenotypically and molecularly. Although the standard treatments have resulted in clinical responses in a subset of patients, though, many patients relapse and no longer respond to further treatments. Hence, both the innate and adaptive resistance to treatments are the main challenges in today's treatment strategies. Noteworthy, several novel treatment strategies, particularly immunotherapies, used alone or in combination, have been developed and that have significantly improved the therapeutic response of many unresponsive cancer patients. Nevertheless, even with the latest new developments of therapeutics that were effective in a larger subset of patients, there is still an urgent need to treat the remaining unresponsive subset of patients. This requires the development of new targeting agents of superior antitumor activities that will lead to overcoming the unaffected resistance by current treatments. There has been accumulating evidence suggesting nitric oxide donors as such targeting agents and considering their pleiotropic antitumor activities, including both the reversal of chemo and immuno-resistance of various unresponsive resistant cancers. The in vitro and in vivo preclinical findings corroborate the sensitizing antitumor activities of nitric oxide donors. In addition, a few clinical findings with NO donors that have been applied in patients have corroborated their antitumor and sensitizing activities in combination with standard therapies. In this review, the role and underlying mechanisms by which nitric oxide donors sensitize cancer resistant cells to both chemotherapy and immunotherapy are briefly described.
Collapse
Affiliation(s)
- Benjamin Bonavida
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, United States.
| |
Collapse
|
8
|
Das D, Fayazzadeh E, Li X, Koirala N, Wadera A, Lang M, Zernic M, Panick C, Nesbitt P, McLennan G. Quiescent hepatic stellate cells induce toxicity and sensitivity to doxorubicin in cancer cells through a caspase-independent cell death pathway: Central role of apoptosis-inducing factor. J Cell Physiol 2020; 235:6167-6182. [PMID: 31975386 DOI: 10.1002/jcp.29545] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/03/2020] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma (HCC) is a major health problem worldwide and in the United States as its incidence has increased substantially within the past two decades. HCC therapy remains a challenge, primarily due to underlying liver disorders such as cirrhosis that determines treatment approach and efficacy. Activated hepatic stellate cells (A-HSCs) are the key cell types involved in hepatic fibrosis/cirrhosis. A-HSCs are important constituents of HCC tumor microenvironment (TME) and support tumor growth, chemotherapy resistance, cancer cell migration, and escaping immune surveillance. This makes A-HSCs an important therapeutic target in hepatic fibrosis/cirrhosis as well as in HCC. Although many studies have reported the role of A-HSCs in cancer generation and investigated the therapeutic potential of A-HSCs reversion in cancer arrest, not much is known about inactivated or quiescent HSCs (Q-HSCs) in cancer growth or arrest. Here we report that Q-HSCs resist cancer cell growth by inducing cytotoxicity and enhancing chemotherapy sensitivity. We observed that the conditioned media from Q-HSCs (Q-HSCCM) induces cancer cell death through a caspase-independent mechanism that involves an increase in apoptosis-inducing factor expression, nuclear localization, DNA fragmentation, and cell death. We further observed that Q-HSCCM enhanced the efficiency of doxorubicin, as measured by cell viability assay. Exosomes present in the conditioned media were not involved in the mechanism, which suggests the role of other factors (proteins, metabolites, or microRNA) secreted by the cells. Identification and characterization of these factors are important in the development of effective HCC therapy.
Collapse
Affiliation(s)
- Dola Das
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Ehsan Fayazzadeh
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Section of Vascular and Interventional Radiology, Department of Diagnostic Radiology, Imaging Institute, Cleveland Clinic, Cleveland, Ohio
| | - Xin Li
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nischal Koirala
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio
| | - Akshay Wadera
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,School of Medicine, New York Medical College, Valhalla, New York
| | - Min Lang
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maximilian Zernic
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Catherine Panick
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Dotter Department of Interventional Radiology, Oregon Health and Science University, Portland, Oregon
| | - Pete Nesbitt
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Erie, Pennsylvania
| | - Gordon McLennan
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Section of Vascular and Interventional Radiology, Department of Diagnostic Radiology, Imaging Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
9
|
Bouaouiche S, Magadoux L, Dondaine L, Reveneau S, Isambert N, Bettaieb A, Jeannin JF, Laurens V, Plenchette S. Glyceryl trinitrate‑induced cytotoxicity of docetaxel‑resistant prostatic cancer cells is associated with differential regulation of clusterin. Int J Oncol 2019; 54:1446-1456. [PMID: 30720069 DOI: 10.3892/ijo.2019.4708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/21/2018] [Indexed: 11/05/2022] Open
Abstract
Metastatic castration resistant prostate cancer (mCRPC) relapse due to acquired resistance to chemotherapy, such as docetaxel, remains a major threat to patient survival. Resistance of mCRPC to docetaxel can be associated with elevated levels of soluble clusterin (sCLU) and growth differentiation factor‑15 (GDF‑15). Any strategies aiming to modulate sCLU and/or GDF‑15 in docetaxel‑resistant prostate cancer cells present a therapeutic interest. The present study reports the cytotoxic effect of a nitric oxide donor, glyceryl trinitrate (GTN), on docetaxel‑resistant mCRPC human cell lines and demonstrates that GTN displays greater inhibition of cell viability toward docetaxel‑resistant mCRPC cells than on mCRPC cells. It is also demonstrated that GTN modulates the level of expression of clusterin (CLU) which is dependent of GDF‑15, two markers associated with docetaxel resistance in prostate cancer. The results indicate that GTN represses the level of expression of the cytoprotective isoform of CLU (sCLU) and can increase the level of expression of the cytotoxic isoform (nuclear CLU) in docetaxel resistant cells. Furthermore, it was observed that GTN differentially regulates the level of the precursor form of GDF‑15 between resistant and parental cells, and that recombinant GDF‑15 can modulate the expression of CLU isoforms and counteract GTN‑induced cytotoxicity in resistant cells. A link was established between GDF‑15 and the expression of CLU isoforms. The present study thus revealed GTN as a potential therapeutic strategy to overcome docetaxel‑resistant mCRPC.
Collapse
Affiliation(s)
- Sarra Bouaouiche
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Lea Magadoux
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Lucile Dondaine
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Sylvie Reveneau
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | | | - Ali Bettaieb
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Jean-François Jeannin
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Veronique Laurens
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Stephanie Plenchette
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| |
Collapse
|
10
|
Wang J, Bi Y, Ruan H, Sun G, Cui X, Yang X, Qin C. Hollow S-nitrosothiols nanoparticle with polymeric brushes for nitric oxide (NO)-releasing as tumor targeted chemotherapy. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2019; 30:122-136. [PMID: 30522414 DOI: 10.1080/09205063.2018.1556852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A kind of tumor targeting nitric oxide donor nanoparticle with brushes is described in this paper. The poly(4-vinylphenylboronic acid) polymeric brush, which shows glucose and pH dual responsiveness, endows the ability of hollow S-nitrosothiols nanoparticle to accurate recognition and binding with the sialic acid over-expressed type tumor cells, such as HepG2 and MCF-7 cells. In vitro experiments, including cells capture and release experiments, confocal fluorescence microscope characterization, cytotoxicity assay with different cells, demonstrate the selective recognition and the controlled NO release to kill tumor cells for these S-nitrosothiols nanoparticles. Low concentration of the released NO from the S-nitrosothiols nanoparticles in the transmission would participate physiological activity and avoid serious side effects because the endogenous nature and the physiological necessity to regulate normal biological functions. To the best of our knowledge, this is the first report about polymer nanoparticles as NO donors with functional brushes to selectively identify tumor cells and release NO in a controlled manner.
Collapse
Affiliation(s)
- Jilan Wang
- a Department of Anesthesiology , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , P. R. China
| | - Ye Bi
- b Department of Endocrinology , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , P. R. China
| | - Hongyan Ruan
- a Department of Anesthesiology , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , P. R. China
| | - Guoqing Sun
- a Department of Anesthesiology , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , P. R. China
| | - Xianping Cui
- c Department of Hepatobiliary Surgery , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , P. R. China
| | - Xinlin Yang
- d Key Laboratory of Functional Polymer Materials, Ministry of Education , Institute of Polymer Chemistry, College of Chemistry, Nankai University , Tianjin , P. R. China
| | - Chengkun Qin
- c Department of Hepatobiliary Surgery , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , P. R. China
| |
Collapse
|
11
|
de Oliveira GA, Cheng RYS, Ridnour LA, Basudhar D, Somasundaram V, McVicar DW, Monteiro HP, Wink DA. Inducible Nitric Oxide Synthase in the Carcinogenesis of Gastrointestinal Cancers. Antioxid Redox Signal 2017; 26:1059-1077. [PMID: 27494631 PMCID: PMC5488308 DOI: 10.1089/ars.2016.6850] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIGNIFICANCE Gastrointestinal (GI) cancer taken together constitutes one of the most common cancers worldwide with a broad range of etiological mechanisms. In this review, we have examined the impact of nitric oxide (NO) on the etiology of colon, colorectal, gastric, esophageal, and liver cancers. Recent Advances: Despite differences in etiology, initiation, and progression, chronic inflammation has been shown to be a common element within these cancers showing interactions of numerous pathways. NO generated at the inflammatory site contributes to the initiation and progression of disease. The amount of NO generated, time, and site vary and are an important determinant of the biological effects initiated. Among the nitric oxide synthase enzymes, the inducible isoform has the most diverse range, participating in numerous carcinogenic processes. There is emerging evidence showing that inducible nitric oxide synthase (NOS2) plays a central role in the process of tumor initiation and/or development. CRITICAL ISSUES Redox inflammation through NOS2 and cyclooxygenase-2 participates in driving the mechanisms of initiation and progression in GI cancers. FUTURE DIRECTIONS Understanding the underlying mechanism involved in NOS2 activation can provide new insights into important prevention and treatment strategies. Antioxid. Redox Signal. 26, 1059-1077.
Collapse
Affiliation(s)
- Graciele Almeida de Oliveira
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| | - Robert Y S Cheng
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| | - Lisa A Ridnour
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| | - Debashree Basudhar
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| | - Veena Somasundaram
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| | - Daniel W McVicar
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| | - Hugo Pequeno Monteiro
- 2 Laboratório de Sinalização Celular, Universidade Federal de São Paulo , São Paulo, Brazil
| | - David A Wink
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| |
Collapse
|
12
|
Girotti AW. Modulation of the Anti-Tumor Efficacy of Photodynamic Therapy by Nitric Oxide. Cancers (Basel) 2016; 8:E96. [PMID: 27775600 PMCID: PMC5082386 DOI: 10.3390/cancers8100096] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/12/2016] [Accepted: 10/14/2016] [Indexed: 12/16/2022] Open
Abstract
Nitric oxide (NO) produced by nitric oxide synthase (NOS) enzymes is a free radical molecule involved in a wide variety of normophysiologic and pathophysiologic processes. Included in the latter category are cancer promotion, progression, and resistance to therapeutic intervention. Animal tumor photodynamic therapy (PDT) studies several years ago revealed that endogenous NO can reduce PDT efficacy and that NOS inhibitors can alleviate this. Until relatively recently, little else was known about this anti-PDT effect of NO, including: (a) the underlying mechanisms; (b) type(s) of NOS involved; and (c) whether active NO was generated in vascular cells, tumor cells, or both. In addressing these questions for various cancer cell lines exposed to PDT-like conditions, the author's group has made several novel findings, including: (i) exogenous NO can scavenge lipid-derived free radicals arising from photostress, thereby protecting cells from membrane-damaging chain peroxidation; (ii) cancer cells can upregulate inducible NOS (iNOS) after a PDT-like challenge and the resulting NO can signal for resistance to photokilling; (iii) photostress-surviving cells with elevated iNOS/NO proliferate and migrate/invade more aggressively; and (iv) NO produced by photostress-targeted cells can induce greater aggressiveness in non-targeted bystander cells. In this article, the author briefly discusses these various means by which NO can interfere with PDT and how this may be mitigated by use of NOS inhibitors as PDT adjuvants.
Collapse
Affiliation(s)
- Albert W Girotti
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
13
|
Plenchette S, Romagny S, Laurens V, Bettaieb A. [NO and cancer: itinerary of a double agent]. Med Sci (Paris) 2016; 32:625-33. [PMID: 27406774 DOI: 10.1051/medsci/20163206027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Protein S-nitrosylation is now recognized as a ubiquitous regulatory mechanism. Like any post-translational modifications, S-nitrosylation is critical for the control of numerous cellular processes. It is now clear that S-nitrosylation is playing a double game, enhancing or inhibiting the tumor growth or the induction of cell death. Thanks to research aimed at demonstrating NO cytotoxic effects, new therapeutic strategies based on NO donor drugs have emerged. Although therapeutic NO donors can target a large number of proteins, the cellular mechanism is still not fully understood. This review reflects the current state of knowledge on S-nitrosylated proteins that take part of the oncogenic and apoptotic signaling, putting forward proteins with potential interest in cancer therapy.
Collapse
Affiliation(s)
- Stéphanie Plenchette
- Université de Bourgogne Franche-Comté, LIIC EA7269, 7, boulevard Jeanne d'Arc, F-21000 Dijon, France - EPHE, PSL Research University, F-75014 Paris, France
| | - Sabrina Romagny
- Université de Bourgogne Franche-Comté, LIIC EA7269, 7, boulevard Jeanne d'Arc, F-21000 Dijon, France - EPHE, PSL Research University, F-75014 Paris, France
| | - Véronique Laurens
- Université de Bourgogne Franche-Comté, LIIC EA7269, 7, boulevard Jeanne d'Arc, F-21000 Dijon, France - EPHE, PSL Research University, F-75014 Paris, France
| | - Ali Bettaieb
- Université de Bourgogne Franche-Comté, LIIC EA7269, 7, boulevard Jeanne d'Arc, F-21000 Dijon, France - EPHE, PSL Research University, F-75014 Paris, France
| |
Collapse
|
14
|
Kim JC, Ha YJ, Tak KH, Roh SA, Kim CW, Kim TW, Kim SK, Kim SY, Cho DH, Kim YS. Complex Behavior of ALDH1A1 and IGFBP1 in Liver Metastasis from a Colorectal Cancer. PLoS One 2016; 11:e0155160. [PMID: 27152521 PMCID: PMC4859559 DOI: 10.1371/journal.pone.0155160] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 04/25/2016] [Indexed: 01/01/2023] Open
Abstract
Using our data set (GSE50760) previously established by RNA sequencing, the present study aimed to identify upregulated genes associated with colorectal cancer (CRC) liver metastasis (CLM) and verify their biological behavior. The potential roles of candidate genes in tumors were assessed using cell proliferation and invasion assays. Tissue samples were collected from 18 CRC patients with synchronous CLM and two CRC cell lines (SW480 and SW620) were used for transfection and cloning. The roles of the genes identified in CLM were verified using immunohistochemistry in 48 nude mice after intrasplenic transplantation of CRC cells. mRNA and protein expression was determined by quantitative real-time reverse transcription polymerase chain reaction and western blot, respectively. Nine genes were initially selected according to the relevance of their molecular function and biological process and, finally, ALDH1A1 and IGFBP1 were chosen based on differential mRNA expression and a positive correlation with protein expression. The overexpression of ALDH1A1 and IGFBP1 significantly and time-dependently decreased cell proliferation (p ≤ 0.001–0.003) and suppressed invasiveness by ≥3-fold over control cells (p < 0.001) in the SW480 cell line, whereas they had a slight effect on reducing SW620 cell proliferation. The protein expression levels of E-cadherin, N-cadherin, claudin-1, and vimentin were significantly higher in CLM than in primary tumor tissues (p < 0.05). However, the cadherin switch, namely, N-cadherin overexpression with reduced E-cadherin expression, was not observed in CLM tissues and transfected CRC cells. Irrespective of reduced proliferation and invasion found on in vitro cell assays, persistent overexpression of β-catenin, vimentin, and ZO-1 in IGFBP1-overexpressing SW480 cells possibly contributed to CLM development in mice implanted with IGFBP1-overexpressing SW480 cells (CLM occurrences: SW480/IGFBP1-transfected mice vs. SW480/vector- and SW480/ALDH1A1-transfected mice, 4/8 vs. 0/10, p = 0.023). In conclusion, ALDH1A1 and IGFBP1 are differentially overexpressed in CLM and may play a dual role, functioning as both tumor suppressors and metastasis promoters in CRC.
Collapse
Affiliation(s)
- Jin Cheon Kim
- Department of Surgery, University of Ulsan College of Medicine, Seoul, Korea
- Institute of Innovative Cancer Research, Asan Medical Center, Seoul, Korea
- * E-mail: (JCK); (DHC); (YSK)
| | - Ye Jin Ha
- Department of Surgery, University of Ulsan College of Medicine, Seoul, Korea
- Institute of Innovative Cancer Research, Asan Medical Center, Seoul, Korea
| | - Ka Hee Tak
- Department of Surgery, University of Ulsan College of Medicine, Seoul, Korea
- Institute of Innovative Cancer Research, Asan Medical Center, Seoul, Korea
| | - Seon Ae Roh
- Department of Surgery, University of Ulsan College of Medicine, Seoul, Korea
- Institute of Innovative Cancer Research, Asan Medical Center, Seoul, Korea
| | - Chan Wook Kim
- Department of Surgery, University of Ulsan College of Medicine, Seoul, Korea
- Institute of Innovative Cancer Research, Asan Medical Center, Seoul, Korea
| | - Tae Won Kim
- Department of Medicine, University of Ulsan College of Medicine, Seoul, Korea
- Institute of Innovative Cancer Research, Asan Medical Center, Seoul, Korea
| | - Seon-Kyu Kim
- Medical Genomics Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Korea
| | - Seon-Young Kim
- Medical Genomics Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Korea
| | - Dong-Hyung Cho
- Institute of Innovative Cancer Research, Asan Medical Center, Seoul, Korea
- Graduate School of East-West Medical Science, Kyung Hee University, Gyeonggi-do, Korea
- * E-mail: (JCK); (DHC); (YSK)
| | - Yong Sung Kim
- Institute of Innovative Cancer Research, Asan Medical Center, Seoul, Korea
- Medical Genomics Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Korea
- * E-mail: (JCK); (DHC); (YSK)
| |
Collapse
|
15
|
Xu W, Zhao M, Wang Y, Zhu H, Wang Y, Zhao S, Wu J, Peng S. Design, synthesis, and in vivo evaluations of benzyl N ω-nitro-N α-(9H-pyrido[3,4-b]indole-3-carbonyl)- l-argininate as an apoptosis inducer capable of decreasing the serum concentration of P-selectin. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00215c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A series of findings suggest that the discovery of in vivo apoptosis inducers for chemotherapy is of clinical importance.
Collapse
Affiliation(s)
- Wenyun Xu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Haimei Zhu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Yaonan Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Shurui Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| |
Collapse
|
16
|
Munaweera I, Shi Y, Koneru B, Patel A, Dang MH, Di Pasqua AJ, Balkus KJ. Nitric oxide- and cisplatin-releasing silica nanoparticles for use against non-small cell lung cancer. J Inorg Biochem 2015; 153:23-31. [PMID: 26402659 DOI: 10.1016/j.jinorgbio.2015.09.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 09/03/2015] [Accepted: 09/09/2015] [Indexed: 12/20/2022]
Abstract
Nitric oxide (NO) and cisplatin releasing wrinkle-structured amine-modified mesoporous silica (AMS) nanoparticles have been developed for the treatment of non-small cell lung cancer (NSCLC). The AMS and NO- and cisplatin-loaded AMS materials were characterized using TEM, BET surface area, FTIR and ICP-MS, and tested in cell culture. The results show that for NSCLC cell lines (i.e., H596 and A549), the toxicity of NO- and cisplatin-loaded silica nanoparticles (NO-Si-DETA-cisplatin-AMS) is significantly higher than that of silica nanoparticles loaded with only cisplatin (Si-DETA-cisplatin-AMS). In contrast, the toxicity of NO-Si-DETA-cisplatin-AMS toward normal lung cell lines is not significantly different from that of Si-DETA-cisplatin-AMS (normal lung fibroblast cells WI-38) or is even lower than that of Si-DETA-cisplatin-AMS (normal lung epithelial cells BEAS-2B). The NO-induced sensitization of tumor cell death demonstrates that NO is a promising enhancer of platinum-based lung cancer therapy.
Collapse
Affiliation(s)
- Imalka Munaweera
- Department of Chemistry, University of Texas at Dallas, 800 West Campbell Rd., Richardson, TX 75080, United States
| | - Yi Shi
- Depatment of Pharmaceutical Sciences, University of North Texas System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, United States
| | - Bhuvaneswari Koneru
- Depatment of Pharmaceutical Sciences, University of North Texas System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, United States
| | - Amit Patel
- Depatment of Pharmaceutical Sciences, University of North Texas System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, United States
| | - Mai H Dang
- Depatment of Pharmaceutical Sciences, University of North Texas System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, United States
| | - Anthony J Di Pasqua
- Depatment of Pharmaceutical Sciences, University of North Texas System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, United States.
| | - Kenneth J Balkus
- Department of Chemistry, University of Texas at Dallas, 800 West Campbell Rd., Richardson, TX 75080, United States.
| |
Collapse
|
17
|
Bonavida B, Garban H. Nitric oxide-mediated sensitization of resistant tumor cells to apoptosis by chemo-immunotherapeutics. Redox Biol 2015; 6:486-494. [PMID: 26432660 PMCID: PMC4596920 DOI: 10.1016/j.redox.2015.08.013] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/14/2015] [Accepted: 08/17/2015] [Indexed: 11/17/2022] Open
Abstract
The generation of NO by the various NO synthases in normal and malignant tissues is manifested by various biological effects that are involved in the regulation of cell survival, differentiation and cell death. The role of NO in the cytotoxic immune response was first revealed by demonstrating the induction of iNOS in target cells by immune cytokines (e.g. IFN-γ, IL-1, TNF-α, etc.) and resulting in the sensitization of resistant tumor cells to death ligands-induced apoptosis. Endogenous/exogenous NO mediated its immune sensitizing effect by inhibiting NF-κΒ activity and downstream, inactivating the repressor transcription factor YY1, which inhibited both Fas and DR5 expressions. In addition, NO-mediated inhibition of NF-κΒ activity and inhibition downstream of its anti-apoptotic gene targets sensitized the tumor cells to apoptosis by chemotherapeutic drugs. We have identified in tumor cells a dysregulated pro-survival/anti-apoptotic loop consisting of NF-κB/Snail/YY1/RKIP/PTEN and its modification by NO was responsible, in large, for the reversal of chemo and immune resistance and sensitization to apoptotic mechanisms by cytotoxic agents. Moreover, tumor cells treated with exogenous NO donors resulted in the inhibition of NF-κΒ activity via S-nitrosylation of p50 and p65, inhibition of Snail (NF-κΒ target gene), inhibition of transcription repression by S-nitrosylation of YY1 and subsequent inhibition of epithelial-mesenchymal transition (EMT), induction of RKIP (inhibition of the transcription repressor Snail), and induction of PTEN (inhibition of the repressors Snail and YY1). Further, each gene product modified by NO in the loop was involved in chemo-immunosensitization. These above findings demonstrated that NO donors interference in the regulatory circuitry result in chemo-immunosensitization and inhibition of EMT. Overall, these observations suggest the potential anti-tumor therapeutic effect of NO donors in combination with subtoxic chemo-immuno drugs. This combination acts on multiple facets including reversal of chemo-immune resistance, and inhibition of both EMT and metastasis.
Collapse
Affiliation(s)
- Benjamin Bonavida
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA.
| | - Hermes Garban
- NantBioScience, Inc., NantWorks, LLC., California NanoSystems Institute (CnSI) at the University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
18
|
Abstract
Resistance to current therapeutic interventions is a major challenge in the treatment of patients affected by cancer. While nitric oxide (NO) might have proneoplastic properties, it is now clear that at high doses, NO has a role in cancer therapeutics. Either as a single agent or in combination with other antineoplastic compounds, NO might be used to overcome tumor cell resistance to conventional treatments. The following discussion addresses the role of NO in cancer therapeutics and includes a report on the role of NO donors in the area of cancer therapeutics.
Collapse
|
19
|
Illum H, Wang DH, Dowell JE, Hittson WJ, Torrisi JR, Meyer J, Huerta S. Phase I dose escalation trial of nitroglycerin in addition to 5-fluorouracil and radiation therapy for neoadjuvant treatment of operable rectal cancer. Surgery 2015; 158:460-5. [PMID: 25964028 DOI: 10.1016/j.surg.2015.04.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 01/27/2015] [Accepted: 04/10/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Nitric oxide donors decreased cell survival in vitro and tumor load in vivo in models of rectal cancer subjected to ionizing radiation. Nitroglycerin (NTG) transdermal patches, added to chemotherapy, have been shown to improve outcomes in lung cancer patients. METHODS This open-label, nonrandomized, multicohort, dose escalation, phase I trial had a primary endpoint to evaluate the safety, tolerability, feasibility, dose-limiting toxicity and maximum tolerated dose of topical NTG in addition to 5-fluorouracil and radiation therapy for neoadjuvant treatment of locoregionally advanced operable rectal cancer. The secondary endpoint was rate of pathologic complete response (pCR). Patients were assigned to 3 sequential cohorts of escalating dose levels of commercially available NTG patches (0.2, 0.4, and 0.6 mg/h), each cohort was intended to consist of 3 patients. RESULTS Thirteen patients were enrolled in the trial as specified in the dose escalation protocol. They were all male with a median age of 59.4 ± 2.5 (SEM) years. The observed toxicities were mild to moderate and manageable. Four patients developed asymptomatic grade 3 lymphopenia during the chemoradiation that resolved promptly upon completion. One patient had a non-ST segment elevation MI and 1 patient developed diarrhea. None of these toxicities were attributed to NTG except for 1 patient who developed a grade 3 headache. This required an additional group of patients at the same dose and no other patient experienced headaches. pCR was 17%. CONCLUSION NTG patches are well-tolerated and it is feasible to proceed with a phase II trial at the maximum dose examined (0.6 mg/h).
Collapse
Affiliation(s)
- Henrik Illum
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX
| | - David H Wang
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX
| | - Jonathan E Dowell
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX
| | - William J Hittson
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX
| | - John R Torrisi
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX
| | - Jeffrey Meyer
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX
| | - Sergio Huerta
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX.
| |
Collapse
|
20
|
Su CL, Wang YT, Chang MH, Fang K, Chen K. The novel heterocyclic trioxirane [(1,3,5-tris oxiran-2-yl)methyl)-1,3,5-triazinane-2,4,6-trione (TATT)] exhibits a better anticancer effect than platinum-based chemotherapy by induction of apoptosis and curcumin further enhances its chemosensitivity. Cell Biochem Biophys 2014; 68:597-609. [PMID: 24078402 DOI: 10.1007/s12013-013-9752-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The heterocyclic trioxirane compound [1,3,5-tris((oxiran-2-yl)methyl)-1,3,5-triazinane-2,4,6-trione (TATT)] is a synthetic compound which has been used as an experimental anticancer agent in human clinical trials. Curcumin, an active natural compound in turmeric and curry, is an ingredient commonly used in the traditional diet of many Asian countries. In the present study, we observed that TATT exhibited a better anticancer effect on chemoresistant human colorectal cancer HT-29 cells and displayed less cytotoxicity on normal human umbilical vein endothelial cells, compared with FDA-approved anticancer drugs (cisplatin, carboplatin, or oxaliplatin) using MTT assay. TATT also induced a stronger apoptotic effect than that seen with the three studied anticancer drugs, as characterized by externalization of phosphatidylserine using flow cytometry. Administration of caspase 8-specific inhibitor (z-IETD-fmk) and mitochondrial permeability transition pore inhibitor (cyclosporin A) demonstrated that TATT-induced apoptosis proceeded via both extrinsic and intrinsic signaling pathways. It is noteworthy that coadministration of curcumin further significantly increased TATT-induced cytotoxicity, externalization of phosphatidylserine (representing early apoptosis), and the percentages of cells at the sub-G1 phase (representing late apoptosis), producing an additivity and/or synergistic effect, and vice versa. Suppression of nuclear NF-κB was involved in curcumin-enhanced chemosensitivity of TATT. Overall, our data indicate that TATT exerts a chemotherapeutic effect on colorectal cancer cells and coadministration of curcumin enhances the treatment effect of TATT.
Collapse
Affiliation(s)
- Chun-Li Su
- Department of Human Development and Family Studies, National Taiwan Normal University, No. 162, Sec. 1, He-ping East Road, Taipei, 106, Taiwan,
| | | | | | | | | |
Collapse
|
21
|
Bonavida B. Postulated mechanisms of resistance of B-cell non-Hodgkin lymphoma to rituximab treatment regimens: strategies to overcome resistance. Semin Oncol 2014; 41:667-77. [PMID: 25440611 DOI: 10.1053/j.seminoncol.2014.08.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Antibody-mediated immunotherapy has gained significant momentum since 1997 when the US Food and Drug Administration approved the first monoclonal antibody (mAb) for the treatment of B-cell non-Hodgkin lymphoma (B-NHL), namely, rituximab (chimeric anti-CD20 mAb). Subsequently, more than 20 approved mAbs have been in use clinically for the treatment of various cancers and several non-cancer-related diseases. Further, the combination treatment of mAbs with chemotherapy, immunotherapy, proteaosome inhibitors, and other inhibitors has resulted in synergistic anti-tumor activity with significant objective clinical responses. Despite their successful clinical use, the underlying mechanisms of rituximab's in vivo activities remain elusive. Further, it is not clear why a subset of patients is initially unresponsive and many responding patients become refractory and resistant to further treatments; hence, the underlying mechanisms of resistance are not known, Attempts have been made to develop model systems to investigate resistance to mAb therapy with the hope to apply the findings in both the generation of new therapeutics and in their use as new prognostic biomarkers. This review focuses on the development of resistance to rituximab treatments and discusses possible underlying mechanisms of action, postulated mechanisms of resistance in model systems, and suggested means to overcome resistance. Several prior reviews on the subject of rituximab resistance have been published and the present review both complements as well as adds new topics of relevance.
Collapse
Affiliation(s)
- Benjamin Bonavida
- Department of Microbiology, Immunology and Molecular Genetics, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA.
| |
Collapse
|
22
|
Wang L, Yu K, Zhou BB, Su ZH, Gao S, Chu LL, Liu JR. The inhibitory effects of a new cobalt-based polyoxometalate on the growth of human cancer cells. Dalton Trans 2014; 43:6070-6078. [PMID: 24463531 DOI: 10.1039/c3dt53030b] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A new cobalt-based polyoxometalate, (Himi)2[Bi(2)W2(0)O(66)(OH)(4)Co2(H2O)(6)Na(4) (H2O)14] · 17H2O (imi = iminazole) (BWCN) has been synthesized and structurally characterized. The inhibitory activities against selected human cancer lines were also determined in this study. The cell viability and chemoresistance of BWCN on human colon carcinoma HT-29 cells were assessed by MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazoliumbromide), cell morphology changes, a comet assay and western blot analysis. The typical morphologic changes of apoptosis and DNA damage indicated that BWCN could have a distinct proliferation inhibitory effect on cancer cells. BWCN as a chemotherapeutic agent also induced apoptosis on HT-29 cells and showed a significant expression of cleaved-caspase-3. These results suggested that the active site of BWCN is the polymeric anion based on the basic tectonic block {BiW(9)}, and the possible mechanism is related to the interference of DNA synthesis in cancer cells.
Collapse
Affiliation(s)
- Lu Wang
- Key Laboratory of Synthesis of Functional Materials and Green Catalysis Colleges of Heilongjiang Province, School of Chemistry and Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang 150025, the People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
23
|
Zhang L, Pickard K, Jenei V, Bullock MD, Bruce A, Mitter R, Kelly G, Paraskeva C, Strefford J, Primrose J, Thomas GJ, Packham G, Mirnezami AH. miR-153 supports colorectal cancer progression via pleiotropic effects that enhance invasion and chemotherapeutic resistance. Cancer Res 2013; 73:6435-47. [PMID: 23950211 DOI: 10.1158/0008-5472.can-12-3308] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although microRNAs (miRNA) have been broadly studied in cancer, comparatively less is understood about their role in progression. Here we report that miR-153 has a dual role during progression of colorectal cancer by enhancing cellular invasiveness and platinum-based chemotherapy resistance. miRNA profiling revealed that miR-153 was highly expressed in a cellular model of advanced stage colorectal cancer. Its upregulation was also noted in primary human colorectal cancer compared with normal colonic epithelium and in more advanced colorectal cancer stages compared with early stage disease. In colorectal cancer patients followed for 50 months, 21 of 30 patients with high levels of miR-153 had disease progression compared with others in this group with low levels of miR-153. Functional studies revealed that miR-153 upregulation increased colorectal cancer invasiveness and resistance to oxaliplatin and cisplatin both in vitro and in vivo. Mechanistic investigations indicated that miR-153 promoted invasiveness indirectly by inducing matrix metalloprotease enzyme 9 production, whereas drug resistance was mediated directly by inhibiting the Forkhead transcription factor Forkhead box O3a (FOXO3a). In support of the latter finding, we found that levels of miR-153 and FOXO3a were inversely correlated in matched human colorectal cancer specimens. Our findings establish key roles for miR-153 overexpression in colorectal cancer progression, rationalizing therapeutic strategies to target expression of this miRNA for colorectal cancer treatment.
Collapse
Affiliation(s)
- Lei Zhang
- Authors' Affiliations: University of Southampton Cancer Sciences Division, Somers Cancer Research Building; Department of Colorectal Surgery, Southampton University Hospital NHS Trust, Southampton; Bioinformatics Unit, London Research Institute, Cancer Research UK, London; and School of Cellular and Molecular Medicine, University of Bristol, Medical Sciences Building, Bristol, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Rapozzi V, Della Pietra E, Zorzet S, Zacchigna M, Bonavida B, Xodo LE. Nitric oxide-mediated activity in anti-cancer photodynamic therapy. Nitric Oxide 2013; 30:26-35. [DOI: 10.1016/j.niox.2013.01.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 01/08/2013] [Accepted: 01/17/2013] [Indexed: 02/05/2023]
|
25
|
Linifanib (ABT-869) enhances radiosensitivity of head and neck squamous cell carcinoma cells. Oral Oncol 2013; 49:591-7. [PMID: 23490884 DOI: 10.1016/j.oraloncology.2013.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 02/05/2013] [Accepted: 02/14/2013] [Indexed: 11/23/2022]
Abstract
OBJECTIVES Novel targeted therapeutic strategies to overcome radio-resistance of cancer cells traditionally treated with radiation may improve patient survival with the added benefit of reduced systemic toxicity. Herein, we tested the feasibility of Linifanib (ABT-869), a multi-receptor tyrosine kinase inhibitor of members of vascular endothelial growth factor (VEGF) and platelet derived growth factor (PDGF) receptor families, on radio-sensitization of Head and Neck Squamous Cell Carcinoma (HNSCC). MATERIALS AND METHODS UMSCC-22A and UMSCC-22B cells were treated with Linifanib and γ-radiation response was determined. Cell viability, cytotoxicity, apoptosis induction and cell cycle distribution were examined by MTT assay, colony formation assay and flow cytometry. In addition, expression of STAT3 and downstream signaling proteins were assessed using western immunoblotting. RESULTS Treatment with Linifanib resulted in cell growth inhibition, G2/M cell cycle arrest, induction of cell death via apoptosis, reduced phosphorylation of STAT3, which has been linked to radio-resistance, lower expression of cyclin D1, survivin and increased PARP cleavage. In addition, Linifanib overcame the radio-resistance of the cell lines and significantly enhanced radiation-induced cytotoxicity (p<0.05). CONCLUSION These data suggest the possibility of combining targeted therapeutic such as Linifanib with radiation to enhance inhibition of cell growth and apoptosis in HNSCC cells. Thus, it may provide a novel therapeutic strategy and improve efficacy of radiation against HNSCC in the future.
Collapse
|
26
|
Huerta-Yepez S, Baritaki S, Baay-Guzman G, Hernandez-Luna MA, Hernandez-Cueto A, Vega MI, Bonavida B. Contribution of either YY1 or BclXL-induced inhibition by the NO-donor DETANONOate in the reversal of drug resistance, both in vitro and in vivo. YY1 and BclXL are overexpressed in prostate cancer. Nitric Oxide 2013; 29:17-24. [PMID: 23246440 DOI: 10.1016/j.niox.2012.12.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 12/03/2012] [Accepted: 12/03/2012] [Indexed: 11/28/2022]
Abstract
Nitric oxide (NO) donors have been shown to activate or inhibit constitutively-activated survival/anti-apoptotic pathways, such as NF-κB, in cancer cells. We report here that treatment of drug-resistant human prostate carcinoma cell lines with high levels (500-1000 μM) of the NO-donor DETANONOate sensitized the resistant tumor cells to apoptosis by CDDP and the combination was synergistic. We hypothesized that DETANONOate inhibits previously identified NF-κB-regulated resistant factors such as Yin Yang 1 (YY1) and Bcl-2/BclXL. Lysates from tumor cells treated with DETANONOate showed inhibition of YY1 and BclXL expressions. Transfection with either YY1 or BclXL siRNA resulted in the inhibition of both YY1 and BclXL expressions and sensitized the cells to CDDP apoptosis. Mice bearing PC-3 tumor xenografts and treated with the combination of DETANONOate and CDDP resulted in significant inhibition of tumor growth; treatment with single agent alone did not have any effect on tumor growth. Analysis of patients TMA tissues with prostatic cancer revealed higher expression of both YY1 and BclXL as a function of tumor grades and their levels were directly correlated. Thus, both YY1 and BclXL are potential prognostic biomarkers. Overall, the above findings suggest that one mechanism of DETANONOate-induced sensitization of resistant tumor cells to CDDP correlated with the inhibition of NF-κB and its targets YY1 and BclXL. The examination of the combination of NO donors and cytotoxic therapy in the treatment of resistant prostate cancer may be warranted.
Collapse
Affiliation(s)
- Sara Huerta-Yepez
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Previous studies showed that targeted endothelial nitric oxide synthase (eNOS) disruption in mice with femoral artery occlusion does not impede and transgenic eNOS overexpression does not stimulate collateral artery growth after femoral artery occlusion, suggesting that nitric oxide from eNOS does not play a role in arteriogenesis. However, pharmacologic nitric oxide synthase inhibition with L-NAME markedly blocks arteriogenesis, suggestive of an important role of nitric oxide. To solve the paradox, we studied targeted deletion of eNOS and of inducible nitric oxide synthase (iNOS) in mice and found that only iNOS knockout could partially inhibit arteriogenesis. However, the combination of eNOS knockout and treatment with the iNOS inhibitor L-NIL completely abolished arteriogenesis. mRNA transcription studies (reverse transcriptase-polymerase chain reaction) performed on collateral arteries of rats showed that eNOS and especially iNOS (but not neural nitric oxide synthase) become upregulated in shear stress-stimulated collateral vessels, which supports the hypothesis that nitric oxide is necessary for arteriogenesis but that iNOS plays an important part. This was strengthened by the observation that the nitric oxide donor DETA NONOate strongly stimulated collateral artery growth, activated perivascular monocytes, and increased proliferation markers. Shear stress-induced nitric oxide may activate the innate immune system and activate iNOS. In conclusion, arteriogenesis is completely dependent on the presence of nitric oxide, a large part of it coming from mononuclear cells.
Collapse
|
28
|
Huerta S, Hrom J, Gao X, Saha D, Anthony T, Reinhart H, Kapur P. Tissue microarray constructs to predict a response to chemoradiation in rectal cancer. Dig Liver Dis 2010; 42:679-84. [PMID: 20227932 DOI: 10.1016/j.dld.2010.02.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Revised: 01/26/2010] [Accepted: 02/02/2010] [Indexed: 12/11/2022]
Abstract
PURPOSE To identify, using tissue microarray (TMA), an immunohistochemical panel predictive of response to ionizing radiation (IR) in rectal cancer. METHODS TMA constructs were prepared from archived stage II/III rectal tumors and matching adjacent mucosa (n=38) from patients treated with pre-operative chemoradiation. Immunohistochemistry (IHC) was performed for MIB, Cyclin E, p21, p27, p53, survivin, Bcl-2, and BAX. Immunoreactivity along with clinical variables was subjected to univariate and forward stepwise logistic regression analyses. RESULTS Pathological complete response (pCR) was 23.9%. The number of positive lymph nodes obtained in the resected specimen was associated with pCR. Immunoreactivity for MIB (Sn 15%, Sp 65%, OR 0.33), p53 (Sn 3%, Sp 84%, OR 0.16), Bcl-2 (Sn 11%, Sp 74%, OR 0.35), and BAX (Sn 92%, Sp 80%, OR 46) was associated with pathological response (all p's<0.001). Forward stepwise logistic regression analysis demonstrated that MIB was an independent predictor of a response to chemoradiation (p=0.001). CONCLUSIONS A combined panel of mediators of apoptosis alone or combined with clinical factors is a feasible approach that can be applied to rectal tumor biopsies to predict a response to chemoradiation. The most sensitive factor was BAX; while MIB independently predicted a response to chemoradiation.
Collapse
Affiliation(s)
- Sergio Huerta
- Department of Surgery, University of Texas Southwestern Medical Center, United States.
| | | | | | | | | | | | | |
Collapse
|
29
|
Stevens EV, Carpenter AW, Shin JH, Liu J, Der CJ, Schoenfisch MH. Nitric oxide-releasing silica nanoparticle inhibition of ovarian cancer cell growth. Mol Pharm 2010; 7:775-85. [PMID: 20205473 DOI: 10.1021/mp9002865] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although the potent antitumor activity of nitric oxide (NO) supports its promise as an antineoplastic agent, effective and selective delivery and action on tumor and not normal cells remains a limiting factor. Nanoparticle-based delivery of NO has been considered as one approach to overcome these limitations. Therefore, we determined the utility of NO delivery using silica nanoparticles and evaluated their antitumor efficacy against human ovarian tumor and nontumor cells. The NO-releasing nanoparticles exhibited enhanced growth inhibition of ovarian tumor cells when compared to both control nanoparticles and a previously reported small molecule NO donor, PYRRO/NO. In addition, the NO-releasing nanoparticles showed greater inhibition of the anchorage-independent growth of tumor-derived and Ras-transformed ovarian cells. Confocal microscopy analysis revealed that fluorescently labeled NO-releasing nanoparticles entered the cytosol of the cell and localized to late endosomes and lysosomes. Furthermore, we observed a nanoparticle size dependency on efficacy against normal versus transformed ovarian cells. Our study provides the first application of nanoparticle-derived NO as an antitumor therapy and merits future studies examining nanoparticle formulation for in vivo applications.
Collapse
Affiliation(s)
- Ellen V Stevens
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | |
Collapse
|
30
|
Gao X, Saha D, Kapur P, Anthony T, Livingston EH, Huerta S. Radiosensitization of HT-29 cells and xenografts by the nitric oxide donor DETANONOate. J Surg Oncol 2009; 100:149-58. [PMID: 19507186 DOI: 10.1002/jso.21318] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Mechanisms of radioresistance in rectal cancer remain unclear. OBJECTIVES To determine mechanisms of radioresistance in rectal cancer cells and to assess the role of the nitric oxide donor DETANONOate as a radiosensitizing agent. METHODS Survival was determined by clonogenic assays, apoptosis by PARP-1 cleavage, and phenotypic differences by Western blot analysis. SCID mice bearing HT-29 xenografts were treated with ionizing radiation (IR) [2.0 Gy x 5], DETANONOate [0.4 mg/kg i.p.], or combination treatment. RESULTS Colorectal cancer HT-29-p53-null cells were resistant and HCT-116-p53 wild-type cells sensitive to IR, which correlated with cleaved PARP-1. Increased levels of p21 occurred in HCT-116 cells, while Bcl-2 and survivin were elevated in HT-29 cells. Radiosensitization was achieved with a substantial elevation of cleaved PARP-1 in DETANONOate-HT-29-treated versus control cells, which was accompanied by elevation of p21, p27, and BAX, and a concomitant decrease in Bcl-2. SCID mice bearing HT-29 xenografts demonstrated a 37.6%, 51.1%, and 70.1% inhibition in tumor growth in mice receiving IR, DETANONOate, and combination treatment versus control, respectively. CONCLUSIONS Radioresistant HT-29 cells are p53-null and have substantially decreased levels of p21. DETANONOate radiosensitized HT-29 cells in vitro and in vivo by an additive effect in apoptosis.
Collapse
Affiliation(s)
- Xiaohuan Gao
- Department of Surgery, Dallas VA Medical Center, University of Texas Southwestern, Dallas, Texas, USA
| | | | | | | | | | | |
Collapse
|