1
|
Hankins GR, Harris RT. The Opioid Growth Factor in Growth Regulation and Immune Responses in Cancer. ADVANCES IN NEUROBIOLOGY 2024; 35:45-85. [PMID: 38874718 DOI: 10.1007/978-3-031-45493-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
It has become apparent that endogenous opioids act not only as neurotransmitters and neuromodulators, but have multiple functions in the body. Activation of the opioid system by opiate drugs is associated with a risk of cancer development through direct stimulation of tumor cell proliferation and through immunosuppression. In contrast, the endogenous peptide opioid [Met5]-enkephalin, now commonly referred to as Opioid Growth Factor (OGF), negatively regulates cell proliferation in a wide number of cells during development, homeostasis, and neoplasia. This action is mediated through the opioid growth factor receptor, originally designated the zeta (ζ) opioid receptor. Further, contrary to the traditional notion of opiates as immunosuppressive, endogenous OGF has been shown to possess a number of positive immunomodulatory properties and may provide a beneficial effect in cancer by augmenting the activity of cells involved in both innate and acquired immunity. Taken together, the evidence supports consideration of opioid peptides such as OGF as new strategies for cancer therapy.
Collapse
Affiliation(s)
- Gerald R Hankins
- Department of Biology, West Virginia State University, Institute, WV, USA.
| | - Robert T Harris
- Department of Biology, West Virginia State University, Institute, WV, USA
| |
Collapse
|
2
|
Sánchez ML, Rodríguez FD, Coveñas R. Involvement of the Opioid Peptide Family in Cancer Progression. Biomedicines 2023; 11:1993. [PMID: 37509632 PMCID: PMC10377280 DOI: 10.3390/biomedicines11071993] [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: 06/28/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Peptides mediate cancer progression favoring the mitogenesis, migration, and invasion of tumor cells, promoting metastasis and anti-apoptotic mechanisms, and facilitating angiogenesis/lymphangiogenesis. Tumor cells overexpress peptide receptors, crucial targets for developing specific treatments against cancer cells using peptide receptor antagonists and promoting apoptosis in tumor cells. Opioids exert an antitumoral effect, whereas others promote tumor growth and metastasis. This review updates the findings regarding the involvement of opioid peptides (enkephalins, endorphins, and dynorphins) in cancer development. Anticancer therapeutic strategies targeting the opioid peptidergic system and the main research lines to be developed regarding the topic reviewed are suggested. There is much to investigate about opioid peptides and cancer: basic information is scarce, incomplete, or absent in many tumors. This knowledge is crucial since promising anticancer strategies could be developed alone or in combination therapies with chemotherapy/radiotherapy.
Collapse
Affiliation(s)
- Manuel Lisardo Sánchez
- Laboratory of Neuroanatomy of the Peptidergic Systems, Institute of Neurosciences of Castilla and León (INCYL), University of Salamanca, 37007 Salamanca, Spain
| | - Francisco D Rodríguez
- Department of Biochemistry and Molecular Biology, Faculty of Chemical Sciences, University of Salamanca, 37007 Salamanca, Spain
- Group GIR-USAL: BMD (Bases Moleculares del Desarrollo), University of Salamanca, 37007 Salamanca, Spain
| | - Rafael Coveñas
- Laboratory of Neuroanatomy of the Peptidergic Systems, Institute of Neurosciences of Castilla and León (INCYL), University of Salamanca, 37007 Salamanca, Spain
- Group GIR-USAL: BMD (Bases Moleculares del Desarrollo), University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
3
|
Vijayakumar J, Haddad T, Gupta K, Sauers J, Yee D. An open label phase II study of safety and clinical activity of naltrexone for treatment of hormone refractory metastatic breast cancer. Invest New Drugs 2023; 41:70-75. [PMID: 36441436 PMCID: PMC10030534 DOI: 10.1007/s10637-022-01317-4] [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: 02/23/2022] [Accepted: 11/02/2022] [Indexed: 11/30/2022]
Abstract
The opioid receptor (OR) antagonist naltrexone inhibits estrogen receptor-α (ER) function in model systems. The goal of this study was to determine the clinical activity of naltrexone in patients with ER-positive metastatic breast cancer. Patients with hormone receptor positive metastatic breast cancer were enrolled on a phase II study of naltrexone. An escalating dose scheme was used to reach the planned dose of 50 mg daily. The primary objective of the study was to evaluate response to therapy as measured by stabilization or reduction of the tumor Maximum Standardized Uptake Value (SUVmax) at 4 weeks by PET-CT scan. The secondary objectives included safety assessment and tumor SUVmax at 8 weeks. Out of 13 patients we enrolled, 8 patients had serial PET-CT scans that were evaluable for response. Of these 8 patients, 5 had stable or decreased SUVmax values at 4 weeks and 3 had clinical or imaging progression. Median time to progression was short at 7 weeks. Naltrexone was well tolerated. There were no discontinuations due to toxicity and no grade 3 or 4 toxicities were noted. Naltrexone showed modest activity in this short study suggesting the contribution of opioid receptors in ER-positive breast cancer. Our data do not support further development of naltrexone in hormone refractory breast cancer. It is possible that more potent peripherally acting OR antagonists may have a greater effect. (ClinicalTrials.gov Identifier: NCT00379197 September 21, 2006).
Collapse
Affiliation(s)
- Jayanthi Vijayakumar
- Division of Hematology Oncology and Transplantation Department of Medicine, University of Minnesota, MN, Minneapolis, USA
| | | | - Kalpna Gupta
- Division of Hematology Oncology and Transplantation Department of Medicine, University of Minnesota, MN, Minneapolis, USA
- Division of Hematology/Oncology, Department of Medicine, University of California, CA, Irvine, USA
| | - Janet Sauers
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Douglas Yee
- Division of Hematology Oncology and Transplantation Department of Medicine, University of Minnesota, MN, Minneapolis, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- , 420 Delaware Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
4
|
Antagonists of the Mu-Opioid Receptor in the Cancer Patient: Fact or Fiction? Curr Oncol Rep 2022; 24:1337-1349. [PMID: 35648340 PMCID: PMC9474368 DOI: 10.1007/s11912-022-01295-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2022] [Indexed: 12/03/2022]
Abstract
Purpose of Review Antagonists of mu-opioid receptor role in cancer progression remains to be elucidated. The objective of this review was to summarize the available evidence on antagonists of mu-opioid receptor effect on tumor progression and prognosis in different types of cancers and an evaluation of the available findings on their mechanism of action. Recent Findings We have found studies related to methylnaltrexone (MNTX) and naltrexone (NTX) usage in cancer outcomes-related setting. We found consistent preclinical evidence of a potential action of MNTX and NTX on cancer growth and spread mediated mainly by effect on the opioid growth factor receptor (OGFr) axis, which results in depressed cell replication. However, clinical results are scarce and limited to poor-quality evidence. Summary Further high-quality studies are warranted to study antagonists of mu-opioid receptor role as a therapeutic option in different types of cancer, especially in patients where the classical treatment causes unacceptable side effects.
Collapse
|
5
|
Gorur A, Patiño M, Shi T, Corrales G, Takahashi H, Rangel R, Gleber-Netto FO, Pickering C, Myers JN, Cata JP. Low doses of methylnaltrexone inhibits head and neck squamous cell carcinoma growth in vitro and in vivo by acting on the mu-opioid receptor. J Cell Physiol 2021; 236:7698-7710. [PMID: 34038587 DOI: 10.1002/jcp.30421] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 05/05/2021] [Accepted: 05/08/2021] [Indexed: 11/06/2022]
Abstract
The Mu-opioid receptor (MOR) has been implicated in tumorigenesis and metastasis. Methylnaltrexone (MNTX), an antagonist of MOR, has shown to inhibit tumor growth and metastasis in lung cancer cell lines. The effect of MNTX on other cell lines such as head and neck squamous cell carcinoma (HNSCC) has not been investigated. We measured the expression and activity of the receptor in different HNSCC cell lines. Then, we evaluated the impact of modulating the expression MOR and the effect of MNTX on the proliferation, clonogenic activity, invasion, and migration of two HNSCC (FaDu and MDA686Tu) cell lines expressing MOR and one cell line (UMSCC47) not expressing the receptor. We also evaluated the impact of MNTX on tumor growth and metastasis formation in vivo. Activation of the receptor with [d-Ala2,N-Me-Phe4, Gly5-ol] (DAMGO) caused a significant reduction in cyclic adenosine monophosphate levels in FaDu cells. Knockdown of MOR inhibited in vitro aggressive cell behaviors on FaDu and MDA686Tu cells and correlated with a reduction in markers of epithelial-mesenchymal transition. In vitro studies showed that MNTX strongly inhibited the proliferation, clonogenic activity, invasion, and migration of FaDu and MDA686Tu cells but has no effect on UMSCC47 cells. In vivo experiments demonstrated that MNTX suppresses tumor growth in HNSCC cell tumor-bearing mice. Our studies indicate that MOR could be considered as a therapeutic target to treat HNSCC.
Collapse
Affiliation(s)
- Aysegul Gorur
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Anesthesiology and Surgical Oncology Research Group, Houston, Texas, USA
| | - Miguel Patiño
- Department of Anesthesiology, Massachusetts General Hospital, Harvard University, Boston, Massachusetts, USA
| | - Ted Shi
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Anesthesiology and Surgical Oncology Research Group, Houston, Texas, USA
| | - German Corrales
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hideaki Takahashi
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Roberto Rangel
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Frederico O Gleber-Netto
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Curtis Pickering
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey N Myers
- Anesthesiology and Surgical Oncology Research Group, Houston, Texas, USA
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Juan P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Anesthesiology and Surgical Oncology Research Group, Houston, Texas, USA
| |
Collapse
|
6
|
Research progress of opioid growth factor in immune-related diseases and cancer diseases. Int Immunopharmacol 2021; 99:107713. [PMID: 34426103 DOI: 10.1016/j.intimp.2021.107713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022]
Abstract
Methionine enkephalin (MENK) has an important role in both neuroendocrine and immune systems. MENK was known as an opioid growth factor (OGF) for its growth regulatory characteristics. OGF interacts with the OGF receptor (OGFr) to inhibit DNA synthesis by upregulating p16 and/or p21, which delays the cell cycle transition from G0/G1 to S phase, and inhibits cell proliferation. In addition, OGF combines with OGFr in immune cells to exert its immunomodulatory activity and regulate immune function. OGF has been studied as an immunomodulator in a variety of autoimmune diseases, including multiple sclerosis, inflammatory bowel disease, diabetes and viral infections, and has been proven to relieve symptoms of certain diseases in animal and in vitro experiments. Also, OGF and OGFr have various anti-tumor molecular mechanisms. OGF can be used as the primary therapy alone or combined with other drugs to treat tumors. This article summarizes the research progress of OGF in immune-related diseases and cancer diseases.
Collapse
|
7
|
Qu N, Meng Y, Handley MK, Wang C, Shan F. Preclinical and clinical studies into the bioactivity of low-dose naltrexone (LDN) for oncotherapy. Int Immunopharmacol 2021; 96:107714. [PMID: 33989971 DOI: 10.1016/j.intimp.2021.107714] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 12/31/2022]
Abstract
Naltrexone (NTX) is a nonspecific opioid antagonist that exerts pharmacological effects on the opioid axis by blocking opioid receptors distributed in cytoplastic and nuclear regions. NTX has been used in opioid use disorder (OUD), immune-associated diseases, alcoholism, obesity, and chronic pain for decades. However, low-dose naltrexone (LDN) also exhibits remarkable inhibition of DNA synthesis, viability, and other functions in numerous cancers and is involved in immune remodeling against tumor invasion and chemical toxicity. The potential anticancer activity of LDN is a focus of basic research. Herein, we summarize the associated studies on LDN oncotherapy to highlight the potential mechanisms and prospective clinical applications.
Collapse
Affiliation(s)
- Na Qu
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Institute and Hospital, No. 44, Xiaoheyan Road, Shenyang 110042, Liaoning Province, China
| | - Yiming Meng
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Institute and Hospital, No. 44, Xiaoheyan Road, Shenyang 110042, Liaoning Province, China
| | - Mike K Handley
- Cytocom, Inc., 2537 Research Blvd. Suite 201, FortCollins, CO 80526, USA
| | - Chunyan Wang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Institute and Hospital, No. 44, Xiaoheyan Road, Shenyang 110042, Liaoning Province, China.
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, No. 77, Puhe Road, Shenyang 110122, China.
| |
Collapse
|
8
|
|
9
|
|
10
|
Naltrexone's Impact on Cancer Progression and Mortality: A Systematic Review of Studies in Humans, Animal Models, and Cell Cultures. Adv Ther 2021; 38:904-924. [PMID: 33337537 DOI: 10.1007/s12325-020-01591-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 11/28/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Naltrexone (NTX) is an opioid antagonist traditionally used as a treatment for alcohol and opioid use disorders, but various studies have documented its involvement in cancer progression, exploring possible anticancer potential, when administered at high doses or as low dose naltrexone (LDN). Herein we present a systematic review of cancer-related outcomes from case reports, clinical trials, and retrospective and prospective studies conducted using cell cultures, animal models, and human subjects receiving NTX/LDN. METHODS A systematic search of NTX in cancer therapy was conducted. Outcomes including tumor size and number, latency to tumor development, survival duration, progression of disease, and scan results were assessed in clinical and animal studies, and cell number was used as the outcome measure of culture studies. RESULTS Several case reports demonstrate notable survival durations and metastatic resolutions in patients with late stage cancer when administered an average LDN dose of 3-5 mg/day. Animal and cell culture studies suggest an overarching principle of NTX involvement in cancer pharmacophysiology, suggesting that high doses and continuous administration can foster cancer progression, whereas low doses and intermittent treatment may hinder cell proliferation, impede tumorigenesis, and have potential anticancer efficacy. CONCLUSION This review emphasizes the value of potential future research on NTX in cancer therapy, and warrants need for a better understanding of underlying mechanisms. Future controlled studies with more robust sample sizes, particularly in humans, are needed to fully elucidate its potential in cancer therapy.
Collapse
|
11
|
Liu N, Yan L, Shan F, Wang X, Qu N, Handley MK, Ma M. Low-dose naltrexone plays antineoplastic role in cervical cancer progression through suppressing PI3K/AKT/mTOR pathway. Transl Oncol 2021; 14:101028. [PMID: 33540155 PMCID: PMC7859308 DOI: 10.1016/j.tranon.2021.101028] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/05/2021] [Accepted: 01/20/2021] [Indexed: 12/24/2022] Open
Abstract
LDN inhibited proliferation in cervical cancer. LDN inhibited migration and invasion in cervical cancer cells. LDN mediated the propagation property in cervical cancer through PI3K/AKT/mTOR signaling pathway.
The incidence of cervical cancer is increasing annually worldwide. Low-dose naltrexone (LDN) has been reported to delay tumor progression, but the mechanism remains unclear. Here, we found that low-dose naltrexone could upregulate the expression of OGFr. Additionally, LDN could suppress the abilities of colony formation, migration and invasion in cervical cancer cells. LDN could also inhibit cervical cancer progression in mice model. Moreover, LDN indirectly reduced the expressions of PI3K, pAKT and mTOR in vitro and in vivo. Therefore, LDN may be considered a potential treatment option for cervical cancer.
Collapse
Affiliation(s)
- Ning Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Limei Yan
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Fengping Shan
- Department of Immunology, College of Basic Medical Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Xiaonai Wang
- Department of Immunology, College of Basic Medical Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Na Qu
- Department of Gynecology, Cancer hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning, China
| | - Mike K Handley
- Cytocom Inc., 37 North Orange Avenue, Suite 607, Orlando, FL 32801, USA
| | - Mingxing Ma
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China.
| |
Collapse
|
12
|
Opioid receptors beyond pain control: The role in cancer pathology and the debated importance of their pharmacological modulation. Pharmacol Res 2020; 159:104938. [DOI: 10.1016/j.phrs.2020.104938] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/24/2020] [Accepted: 05/15/2020] [Indexed: 12/15/2022]
|
13
|
Tregubenko P, Zvonarev V. Impact of Opioid Use in Hematological Malignancies: Clinical, Immunological and Concomitant Aspects. J Hematol 2020; 9:41-54. [PMID: 32855752 PMCID: PMC7430860 DOI: 10.14740/jh689] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/02/2020] [Indexed: 02/06/2023] Open
Abstract
Opioid agents play a unique role in pain and symptom management for cancer patients. Research shows that opiate use, especially when associated with underlying cancer, has significant effects on hematological parameters. These changes may lead to greater risk for immunosuppression, tumor growth and progression of metastatic processes. The aim of this review is to explore the effects of opiates on various metabolic and biological processes, as well as the hematopoietic system, especially in cancer patients. Our findings demonstrate that the tumor-promoting effects of opiates remain contradictory, as both growth-promoting and anti-tumor effects have been observed. However, available data suggest that opiates can facilitate the proliferation and migration of tumor cells, and understanding of this process on cancer treatment is tremendously important.
Collapse
Affiliation(s)
- Polina Tregubenko
- Internal Medicine Residency Program, Albert Einstein College of Medicine, Jacobi Medical Center, Bronx, NY, USA
| | - Valeriy Zvonarev
- School of Behavioral Sciences, California Southern University, Costa Mesa, CA, USA.,Psychiatry Residency Training Program, Center for Behavioral Medicine, UMKC, 1000 E. 24th Street, Kansas City, MO 64108, USA
| |
Collapse
|
14
|
Liu N, Ma M, Qu N, Wang R, Chen H, Hu F, Gao S, Shan F. Low-dose naltrexone inhibits the epithelial-mesenchymal transition of cervical cancer cells in vitro and effects indirectly on tumor-associated macrophages in vivo. Int Immunopharmacol 2020; 86:106718. [PMID: 32585612 DOI: 10.1016/j.intimp.2020.106718] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/24/2022]
Abstract
The metastasis of cervical cancer has always been a clinical challenge. We investigated the effects of low-dose naltrexone (LDN) on the epithelial mesenchymal transition of cervical cancer cells in vitro as well as its influence on macrophage polarization and associated cytokines in vivo. The results suggested that LDN supressed the proliferation, migration and invasion abilities and promote their apoptosis in Hela cells, whereas the opioid growth factor receptor (OGFr) silenced significantly reversed these effects in vitro. Knockdown the expression of OGFr, the inhibitory of LDN on EMT was weakened. LDN could inhibit cervical cancer progression in nude mice. In additon, LDN indirectly reduced the number of tumor-associated macrophages (TAMs), mainly M2 macrophages, and decreased expression of anti-inflammatory factor IL-10 in the serum of nude mice. These findings demonstrate that LDN could be a potential treatment for cervical cancer.
Collapse
Affiliation(s)
- Ning Liu
- Department of Immunology, School of Basic Medical Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning Province, China; Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, Liaoning Province, China
| | - Mingxing Ma
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning Province, China
| | - Na Qu
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, China
| | - Ruizhe Wang
- Department of Gynecology, No. 1 Affiliated Hospital, China Medical University, No. 155, North Nanjing Street, Heping District, Shenyang 110001, Liaoning, China
| | - Hao Chen
- Department of Immunology, School of Basic Medical Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning Province, China
| | - Fangzhu Hu
- Department of Immunology, School of Basic Medical Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning Province, China
| | - Song Gao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, Liaoning Province, China
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning Province, China.
| |
Collapse
|
15
|
Ma M, Wang X, Liu N, Shan F, Feng Y. Low-dose naltrexone inhibits colorectal cancer progression and promotes apoptosis by increasing M1-type macrophages and activating the Bax/Bcl-2/caspase-3/PARP pathway. Int Immunopharmacol 2020; 83:106388. [DOI: 10.1016/j.intimp.2020.106388] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 03/06/2020] [Indexed: 12/24/2022]
|
16
|
Li G, Li B, Song J, Wang N, Gao Z. Endomorphin-2 Analog Inhibits the Growth of DLD-1 and RKO Human Colon Cancer Cells by Inducing Cell Apoptosis. Med Sci Monit 2020; 26:e921251. [PMID: 32336747 PMCID: PMC7199432 DOI: 10.12659/msm.921251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background In developed countries, colon cancer is a leading cause of cancer-associated mortality. Dietary changes have resulted in an increased incidence of colon cancer in Asia. This study aimed to investigate the effects of the structural analog of endomorphin-2 (H-Tyr-Pro-Phe-Phe-NH2) on human colon cancer cells in vitro. Material/Methods Human DLD-1 and RKO colon cancer cells and CCD-18Co normal human colonic fibroblasts were treated with increasing doses of the structural analog of endomorphin-2. Cells underwent the MTT assay, fluorescence confocal flow cytometry, and Hoechst 33258 staining to investigate cell proliferation, the cell cycle, and apoptosis. Western blot was used to measure the expression levels of poly(ADP-ribose) polymerase-1 (PARP-1), cytochrome c, caspase-3, and caspase-9. The 2′,7′-dichlorofluorescein diacetate (DCFH-DA) fluorescence method measured reactive oxygen species (ROS). Results Cell proliferation of DLD-1 and RKO cells was inhibited by the endomorphin-2 analog in a dose-dependent manner, and a 100 μM dose reduced DLD-1 and RKO cell proliferation by 28% and 23%, respectively, at 72 h. Endomorphin-2 analog induced cell apoptosis and the generation of ROS, activated caspase-3 and caspase-9, and increased the levels of p53 and cytochrome c release, and down-regulated of Akt activation in DLD-1 and RKO cells in a dose-dependent manner. Treatment of the DLD-1 and RKO cells with the endomorphin-2 analog increased the expression of Bax and reduced the expression of Bcl-2. Conclusions Endomorphin-2 analog inhibited colon cancer cell proliferation, activated apoptosis, and down-regulated Akt phosphorylation of human DLD-1 and RKO colon cancer cells in vitro in a dose-dependent manner.
Collapse
Affiliation(s)
- Guanghua Li
- Department of Gastrointestinal Surgery, The Second Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Bo Li
- Department of Electrocardiography, Peoples' Hospital of Zhangqiu, Jinan, Shandong, China (mainland)
| | - Jingang Song
- Department of General Surgery, Dezhou Peoples' Hospital, Dezhou, Shandong, China (mainland)
| | - Na Wang
- Department of Oncology, Peoples' Hospital of Zhangqiu, Jinan, Shandong, China (mainland)
| | - Zhuanglei Gao
- Department of Gastrointestinal Surgery, The Second Hospital of Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
17
|
Aboalsoud A, El-Ghaiesh SH, Abd Elmonem FF, Salem ML, Abdel Rahman MN. The effect of low-dose naltrexone on solid Ehrlich carcinoma in mice: The role of OGFr, BCL2, and immune response. Int Immunopharmacol 2020; 78:106068. [PMID: 31835085 DOI: 10.1016/j.intimp.2019.106068] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/11/2019] [Accepted: 11/19/2019] [Indexed: 02/07/2023]
Abstract
AIMS Cancer is a major worldwide health problem. Cancer cells express opioid growth factor (OGF) which controls their growth. Naltrexone in low dose (LDN) blocks opioid receptors intermittently and controls the replication of cancer cells. The aim of this study was to investigate the effect of LDN and its chemotherapeutic additive effect on the growth of solid Ehrlich carcinoma in mice with focus on the OGFr and immune responses. MAIN METHODS Sixty female Swiss albino mice were assigned into 5 groups (n: 12 mice each): (i): normal control, (ii): Solid Ehrlich carcinoma (SEC), (iii): SEC treated with LDN, (iv): SEC treated with 5-fluorouracil (5-FU), (v): SEC treated with LDN + 5-FU. All drugs were started when the tumor became palpable on 9th day. At the end of the study animals were sacrificed, blood and tissue samples were collected. Tumor weight and volume were measured. Splenocytes and myeloid derived suppressor cells (MDSC) were counted. Tumor expression of opioid growth factor receptors (OGFr), serum level of IFN-γ, tumor histopathology (H&E) and immunohistochemistry staining of p21, p53, Bcl2 were assessed. KEY FINDINGS All drug-treated groups showed reduction in tumor weight and volume, significant increase of splenocyte with tendency to reduce MDSC cell counts. LDN led to significant increase in OGFr both in solo and in combination with 5FU. Serum IFN-γ is significantly increased by LDN but decreased by 5-FU. Also, LDN and 5FU increased immunehistochemical staining of p21 while decreased immunostaining of Bcl2. In animals treated with a combination of LDN and 5FU a maximal downregulation of the antiapoptotic mediator BCL2 was observed. SIGNIFICANCE The current study suggested that LDN may play a role in inhibiting cancer cell growth and highlights the possibility of promising combination with cancer chemotherapeutics, which guarantee further clinical studies for approval.
Collapse
Affiliation(s)
- Alshimaa Aboalsoud
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Sabah H El-Ghaiesh
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt; Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia.
| | - Fleur F Abd Elmonem
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mohammed L Salem
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mohamed N Abdel Rahman
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt; Department of Clinical Pharmacology, Faculty of Medicine, Bisha University, Bisha, Saudi Arabia
| |
Collapse
|
18
|
Effect of low dose naloxone on the immune system function of a patient undergoing video-assisted thoracoscopic resection of lung cancer with sufentanil controlled analgesia - a randomized controlled trial. BMC Anesthesiol 2019; 19:236. [PMID: 31856760 PMCID: PMC6923917 DOI: 10.1186/s12871-019-0912-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/09/2019] [Indexed: 01/29/2023] Open
Abstract
Background Perioperative immune function plays an important role in the prognosis of patients. Several studies have indicated that low-dose opioid receptor blockers can improve immune function. Methods Sixty-nine patients undergoing video-assisted thoracoscopic resection of the lung cancer were randomly assigned to either the naloxone group (n = 35) or the non-naloxone group (n = 34) for postoperative analgesia during the first 48 h after the operation. Both groups received sufentanil and palonosetron via postoperative analgesia pump, while 0.05 μg·kg− 1·h− 1 naloxone was added in naloxone group. The primary outcomes were the level of opioid growth factor (OGF) and immune function assessed by natural killer cells and CD4+/CD8+ T-cell ratio. Second outcomes were assessed by the intensity of postoperative pain, postoperative rescue analgesia dose, postoperative nausea and vomiting (PONV). Results The level of OGF in the naloxone group increased significantly at 24 h (p<0.001) and 48 h after the operation (P < 0.01). The natural killer cells (P < 0.05) and CD4+/CD8+ T-cell ratio (P < 0.01) in the naloxone group increased significantly at 48 h after the operation. The rest VAS scores were better with naloxone at 12 and 24 h after operation(P < 0.05), and the coughing VAS scores were better with naloxone at 48 h after the operation(P < 0.05). The consumption of postoperative rescue analgesics in the naloxone group was lower (0.00(0.00–0.00) vs 25.00(0.00–62.50)), P < 0.05). Postoperative nausea scores at 24 h after operation decreased in naloxone group(0.00 (0.00–0.00) vs 1.00 (0.00–2.00), P < 0.01). Conclusion Infusion of 0.05 μg·kg− 1·h− 1 naloxone for patients undergoing sufentanil-controlled analgesia for postoperative pain can significantly increase the level of OGF, natural killer cells, and CD4+/CD8+ T-cell ratio compared with non-naloxone group, and postoperative pain intensity, request for rescue analgesics, and opioid-related side effects can also be reduced. Trial registration The trial was registered at the Chinese Clinical Trial Registry on January 26, 2019 (ChiCTR1900021043).
Collapse
|
19
|
Qu N, Wang X, Meng Y, Shan F. Prospective oncotarget for gynecological cancer: Opioid growth factor (OGF) - opioid growth factor receptor (OGFr) axis. Int Immunopharmacol 2019; 75:105723. [PMID: 31408839 DOI: 10.1016/j.intimp.2019.105723] [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: 06/19/2019] [Accepted: 06/24/2019] [Indexed: 02/07/2023]
Abstract
The standard treatments for neoplasia include surgery, chemotherapy, hormone antagonists and radiotherapy, which can prolong survival, but rarely cure the tumors of gynecological cancer patients. OGF - OGFr expression, in various gynecologic cells and tissues, is an intersection point between cell development, neuroendocrine function and immune modulation. It has been identified that OGF and OGFr expression differs between gynecological tumor and normal cells. Further, exogenous or endogenous OGF and OGFr antagonists have been known to have a role in regulating cell viability and apoptosis. Moreover, the expression of proteins in the OGF - OGFr axis modulate differentiation and membrane expression of immune cells, which can enhance the immune response. In vivo and in vitro assays have shown that OGF and OGFr antagonists inhibit mitosis as well as induce apoptosis in gynecologic cancer cells. Although immune augmentation combination therapies can intensify cytotoxic activity, OGF or OGFr antagonists do not increase toxicities associated with dual-immune regulation. In conclusion, the OGF - OGFr axis provides significant strategies for antitumor efficiency in gynecological cancer.
Collapse
Affiliation(s)
- Na Qu
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Shenyang 110042, Liaoning Province, China
| | - Xiaobin Wang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Shenyang 110042, Liaoning Province, China
| | - Yiming Meng
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Shenyang 110042, Liaoning Province, China
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, No. 77, Puhe Road, Shenyang 110122, China.
| |
Collapse
|
20
|
Machado MC, da Costa-Neto JM, Portela RD, D'Assis MJMH, Martins-Filho OA, Barrouin-Melo SM, Borges NF, Silva FL, Estrela-Lima A. The effect of naltrexone as a carboplatin chemotherapy-associated drug on the immune response, quality of life and survival of dogs with mammary carcinoma. PLoS One 2018; 13:e0204830. [PMID: 30286124 PMCID: PMC6171873 DOI: 10.1371/journal.pone.0204830] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 09/14/2018] [Indexed: 02/02/2023] Open
Abstract
The objective of this study was to evaluate the effect of low-dose naltrexone (LDN) as a carboplatin chemotherapy-associated drug in female dogs with mammary carcinoma in benign mixed tumors (MC-BMT) after mastectomy and to assess its association with quality of life and survival rates. Sixty female dogs were included in this study, all of which had histopathological diagnosis of MC-BMT and were divided into three groups: G1 (control), consisting of animals submitted only to mastectomy with or without regional metastasis; G2, composed of treated animals that did not present with metastasis; and G3, treated dogs that presented with metastasis. G2 and G3 were also subdivided according to the treatment administered: chemotherapy alone (MC-BMT(-) C/MC-BMT(+) C) or LDN and chemotherapy (MC-BMT(-) C+LDN/MC-BMT(+) C+LDN). All animals were subjected to clinical evaluation, mastectomy, peripheral blood lymphocyte immunophenotyping, beta-endorphin and met-enkephalin quantification, and evaluation of survival rates and quality of life scores. The results showed higher serum concentrations of beta-endorphin and met-enkephalin, fewer chemotherapy-related side effects, and better quality of life and survival rates in the LDN-treated groups than in LDN-untreated groups (P < 0.05). Evaluation of clinical and pathological parameters indicated a significant association between the use of LDN and both prolonged survival and enhanced quality of life. These results indicate that LDN is a viable chemotherapy-associated treatment in female dogs with MC-BMT, maintaining their quality of life and prolonging survival rates.
Collapse
Affiliation(s)
- Marília Carneiro Machado
- Department of Anatomy, Pathology and Veterinary Clinics, Federal University of Bahia, Salvador, Bahia, Brazil
| | - João Moreira da Costa-Neto
- Department of Anatomy, Pathology and Veterinary Clinics, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Ricardo Dias Portela
- Laboratory of Immunology and Molecular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | | | - Olindo Assis Martins-Filho
- Laboratory of Diagnostic and Monitoring Biomarkers, Research Center René Rachou, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Stella Maria Barrouin-Melo
- Department of Anatomy, Pathology and Veterinary Clinics, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Natalie Ferreira Borges
- Center for Agrarian, Environmental and Biological Sciences, Federal University of Recôncavo da Bahia, Cruz das Almas, Bahia, Brazil
| | - Fabiana Lessa Silva
- Department of Agrarian and Environmental Sciences, Estadual University of Santa Cruz, Ilhéus, Bahia, Brazil
| | - Alessandra Estrela-Lima
- Department of Anatomy, Pathology and Veterinary Clinics, Federal University of Bahia, Salvador, Bahia, Brazil
- * E-mail:
| |
Collapse
|
21
|
Li Z, You Y, Griffin N, Feng J, Shan F. Low-dose naltrexone (LDN): A promising treatment in immune-related diseases and cancer therapy. Int Immunopharmacol 2018; 61:178-184. [DOI: 10.1016/j.intimp.2018.05.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/22/2018] [Accepted: 05/22/2018] [Indexed: 12/16/2022]
|
22
|
Stockdale DP, Titunick MB, Biegler JM, Reed JL, Hartung AM, Wiemer DF, McLaughlin PJ, Neighbors JD. Selective opioid growth factor receptor antagonists based on a stilbene isostere. Bioorg Med Chem 2017; 25:4464-4474. [PMID: 28693915 PMCID: PMC5567982 DOI: 10.1016/j.bmc.2017.06.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/13/2017] [Accepted: 06/21/2017] [Indexed: 02/01/2023]
Abstract
As part of an ongoing drug development effort aimed at selective opioid receptor ligands based on the pawhuskin natural products we have synthesized a small set of amide isosteres. These amides were centered on lead compounds which are selective antagonists for the delta and kappa opioid receptors. The amide isomers revealed here show dramatically different activity from the parent stilbene compounds. Three of the isomers synthesized showed antagonist activity for the opioid growth factor (OGF)/opioid growth factor receptor (OGFR) axis which is involved in cellular and organ growth control. This cellular signaling mechanism is targeted by "low-dose" naltrexone therapy which is being tested clinically for multiple sclerosis, Crohn's disease, cancer, and wound healing disorders. The compounds described here are the first selective small molecule ligands for the OGF/OGFR system and will serve as important leads and probes for further study.
Collapse
Affiliation(s)
- David P Stockdale
- Department of Chemistry, The University of Iowa, Iowa City, IA 52242-1294, United States
| | - Michelle B Titunick
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Jessica M Biegler
- Department of Pharmacology and Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Pennsylvania State University Cancer Institute, Hershey, PA 17033, United States
| | - Jessie L Reed
- Department of Pharmacology and Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Pennsylvania State University Cancer Institute, Hershey, PA 17033, United States
| | - Alyssa M Hartung
- Department of Chemistry, The University of Iowa, Iowa City, IA 52242-1294, United States
| | - David F Wiemer
- Department of Chemistry, The University of Iowa, Iowa City, IA 52242-1294, United States
| | - Patricia J McLaughlin
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Pennsylvania State University Cancer Institute, Hershey, PA 17033, United States
| | - Jeffrey D Neighbors
- Department of Pharmacology and Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Pennsylvania State University Cancer Institute, Hershey, PA 17033, United States.
| |
Collapse
|
23
|
Cant R, Dalgleish AG, Allen RL. Naltrexone Inhibits IL-6 and TNFα Production in Human Immune Cell Subsets following Stimulation with Ligands for Intracellular Toll-Like Receptors. Front Immunol 2017; 8:809. [PMID: 28744288 PMCID: PMC5504148 DOI: 10.3389/fimmu.2017.00809] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/26/2017] [Indexed: 12/20/2022] Open
Abstract
The opioid antagonist naltrexone hydrochloride has been suggested to be a potential therapy at low dosage for multiple inflammatory conditions and cancers. Little is known about the immune-modulating effects of naltrexone, but an effect on the activity of toll-like receptor 4 (TLR4) has been reported. We analyzed the effects of naltrexone hydrochloride on IL-6 secretion by peripheral blood mononuclear cells (PBMC) in vitro following stimulation with ligands for TLR4 and for the intracellular receptors TLR7, TLR8, and TLR9. Naltrexone did not affect cell viability or induce apoptosis of PBMC. Intracellular staining demonstrated that naltrexone inhibited production of IL-6 and TNFα by monocyte and plasmacytoid dendritic cell subsets within the PBMC population following treatment with ligands for TLR7/8 and TLR9, respectively. No effect of cytokine production by PBMC following stimulation of TLR4 was observed. Additionally, naltrexone inhibited IL-6 production in isolated monocytes and B cells after TLR7/8 and TLR9 stimulation, respectively, but no effect on IL-6 production in isolated monocytes after TLR4 stimulation was observed. These findings indicate that naltrexone has the potential to modulate the secretion of inflammatory cytokines in response to intracellular TLR activity, supporting the hypothesis that it may have potential for use as an immunomodulator.
Collapse
Affiliation(s)
- Rachel Cant
- Institute for Infection and Immunity, St George's University of London, London, United Kingdom
| | - Angus G Dalgleish
- Institute for Infection and Immunity, St George's University of London, London, United Kingdom
| | - Rachel L Allen
- Institute for Infection and Immunity, St George's University of London, London, United Kingdom
| |
Collapse
|
24
|
Ayyagari VN, Hsieh THJ, Diaz-Sylvester PL, Brard L. Evaluation of the cytotoxicity of the Bithionol - cisplatin combination in a panel of human ovarian cancer cell lines. BMC Cancer 2017; 17:49. [PMID: 28086831 PMCID: PMC5234112 DOI: 10.1186/s12885-016-3034-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/15/2016] [Indexed: 11/23/2022] Open
Abstract
Background Combination drug therapy appears a promising approach to overcome drug resistance and reduce drug-related toxicities in ovarian cancer treatments. In this in vitro study, we evaluated the antitumor efficacy of cisplatin in combination with Bithionol (BT) against a panel of ovarian cancer cell lines with special focus on cisplatin-sensitive and cisplatin-resistant cell lines. The primary objectives of this study are to determine the nature of the interactions between BT and cisplatin and to understand the mechanism(s) of action of BT-cisplatin combination. Methods The cytotoxic effects of drugs either alone or in combination were evaluated using presto-blue assay. Cellular reactive oxygen species were measured by flow cytometry. Immunoblot analysis was carried out to investigate changes in levels of cleaved PARP, XIAP, bcl-2, bcl-xL, p21 and p27. Luminescent and colorimetric assays were used to test caspases 3/7 and ATX activity. Results The efficacy of the BT-cisplatin combination depends upon the cell type and concentrations of cisplatin and BT. In cisplatin-sensitive cell lines, BT and cisplatin were mostly antagonistic except when used at low concentrations, where synergy was observed. In contrast, in cisplatin-resistant cells, BT-cisplatin combination treatment displayed synergistic effects at most of the drug ratios/concentrations. Our results further revealed that the synergistic interaction was linked to increased reactive oxygen species generation and apoptosis. Enhanced apoptosis was correlated with loss of pro-survival factors (XIAP, bcl-2, bcl-xL), expression of pro-apoptotic markers (caspases 3/7, PARP cleavage) and enhanced cell cycle regulators p21 and p27. Conclusion In cisplatin-resistant cell lines, BT potentiated cisplatin-induced cytotoxicity at most drug ratios via enhanced ROS generation and modulation of key regulators of apoptosis. Low doses of BT and cisplatin enhanced efficiency of cisplatin treatment in all the ovarian cancer cell lines tested. Our results suggest that novel combinations such as BT and cisplatin might be an attractive therapeutic approach to enhance ovarian cancer chemosensitivity. Combining low doses of cisplatin with subtherapeutic doses of BT can ultimately lead to the development of an innovative combination therapy to reduce/prevent the side effects normally occurring when high doses of cisplatin are administered. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-3034-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vijayalakshmi N Ayyagari
- Division of Gynecologic Oncology; Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Tsung-Han Jeff Hsieh
- Division of Gynecologic Oncology; Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Paula L Diaz-Sylvester
- Division of Gynecologic Oncology; Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, IL, USA.,Center for Clinical Research, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Laurent Brard
- Division of Gynecologic Oncology; Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, IL, USA. .,Simmons Cancer Institute at SIU, Southern Illinois University School of Medicine, Springfield, IL, USA.
| |
Collapse
|
25
|
Yi Z, Guo S, Hu X, Wang X, Zhang X, Griffin N, Shan F. Functional modulation on macrophage by low dose naltrexone (LDN). Int Immunopharmacol 2016; 39:397-402. [DOI: 10.1016/j.intimp.2016.08.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 08/17/2016] [Accepted: 08/17/2016] [Indexed: 12/31/2022]
|
26
|
KREN NANCYP, ZAGON IANS, McLAUGHLIN PATRICIAJ. Mutations in the opioid growth factor receptor in human cancers alter receptor function. Int J Mol Med 2015; 36:289-93. [DOI: 10.3892/ijmm.2015.2221] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/15/2015] [Indexed: 11/06/2022] Open
|
27
|
Meng J, Meng Y, Plotnikoff NP, Youkilis G, Griffin N, Shan F. Low dose naltrexone (LDN) enhances maturation of bone marrow dendritic cells (BMDCs). Int Immunopharmacol 2014; 17:1084-9. [PMID: 24455776 DOI: 10.1016/j.intimp.2013.10.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
It has been demonstrated previously that immune cell activation and proliferation were sensitive to the effects of naltrexone, a non-peptidic δ-opioid receptor selective antagonist and opioid receptors on BMDCs have been detected [1]. However, there is little prior data published on naltrexone and DCs. Therefore, we hypothesized that LDN could exert modulating effect on BMDCs. In present study, we studied influence of LDN on both phenotypic and functional maturation of BMDCs. Changes of BMDC post-treatment with LDN were evaluated using conventional light microscope and transmission electron microscopy (TEM); flow cytometry(FCM); cytochemistry; acid phosphatase activity(ACP) test; FITC-dextran bio-assay; mixed lymphocytes and enzyme-linked immunosorbent assay (ELISA). We have found that LDN enhances maturation of BMDCs as evidenced by 1) up-regulating the expression of MHC II, CD40, CD83, CD80 and CD86 molecules on BMDCs; 2) down-regulating the rates of pinocytosis and phagocytosis accompanied by the results of decreased ACP, and FITC-dextran bio-assay; 3) mounting potential of BMDCs to drive T cell; and 4) inducing secretion of higher levels of IL-12 and TNF-α. It is therefore concluded that LDN can efficiently promote the maturation of BMDCs via precise modulation inside and outside BMDCs. Our study has provided meaningful mode of action on the role of LDN in immunoregulation, and rationale on future application of LDN for enhancing host immunity in cancer therapy and potent use in the design of DC-based vaccines for a number of diseases.
Collapse
|
28
|
Huang Y, Ju B, Tian J, Liu F, Yu H, Xiao H, Liu X, Liu W, Yao Z, Hao Q. Ovarian cancer stem cell-specific gene expression profiling and targeted drug prescreening. Oncol Rep 2014; 31:1235-48. [PMID: 24424387 DOI: 10.3892/or.2014.2976] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 12/23/2013] [Indexed: 01/06/2023] Open
Abstract
Cancer stem cells, with unlimited self-renewal potential and other stem cell characteristics, occur in several types of cancer, including ovarian cancer (OvC). Although CSCs can cause tumor initiation, malignant proliferation, relapse and multi-drug resistance, ways to eliminate them remain unknown. In the present study, we compared ovarian cancer stem cell (OVCSC) expression profiles in normal ovarian surface epithelium and ovarian cells from patients with advanced disease to identify key pathways and specific molecular signatures involved in OVC progression and to prescreen candidate small-molecule compounds with anti-OVCSC activity. Comparison of genome-wide expression profiles of OvC stemness groups with non-stemness controls revealed 6495, 1347 and 509 differentially expressed genes in SDC, SP1 and SP2 groups, respectively, with a cut-off of fold-change set at >1.5 and P<0.05. NAB1 and NPIPL1 were commonly upregulated whereas PROS1, GREB1, KLF9 and MTUS1 were commonly downregulated in all 3 groups. Most differentially expressed genes consistently clustered with molecular functions such as protein receptor binding, kinase activity and chemo-repellent activity. These genes regulate cellular components such as centrosome, plasma membrane receptors, and basal lamina, and may participate in biological processes such as cell cycle regulation, chemoresistance and stemness induction. Key Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways such as ECM receptor, ErbB signaling, endocytosis and adherens junction pathways were enriched. Gene co-expression extrapolation screening by the Connectivity Map revealed several small-molecule compounds (such as SC-560, disulfiram, thapsigargin, esculetin and cinchonine) with potential anti-OVCSC properties targeting OVCSC signature genes. We identified several key CSC features and specific regulation networks in OVCSCs and predicted several small molecules with potential anti-OVCSC pharmacological properties, which may aid the development of OVCSC-specific drugs.
Collapse
Affiliation(s)
- Yuting Huang
- Department of Gynecological Oncology, Tianjin Medical Univerisity Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Baohui Ju
- Department of Gynecological Oncology, Tianjin Medical Univerisity Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Jing Tian
- Department of Gynecological Oncology, Tianjin Medical Univerisity Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Fenghua Liu
- Department of Gynecological Oncology, Tianjin Medical Univerisity Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Hu Yu
- Department of Gynecological Oncology, Tianjin Medical Univerisity Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Huiting Xiao
- Department of Gynecological Oncology, Tianjin Medical Univerisity Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Xiangyu Liu
- Department of Gynecological Oncology, Tianjin Medical Univerisity Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Wenxin Liu
- Department of Gynecological Oncology, Tianjin Medical Univerisity Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Zhi Yao
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Quan Hao
- Department of Gynecological Oncology, Tianjin Medical Univerisity Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| |
Collapse
|
29
|
McLaughlin PJ, Zagon IS. Novel treatment for triple-negative breast and ovarian cancer: endogenous opioid suppression of women's cancers. Expert Rev Anticancer Ther 2014; 14:247-50. [PMID: 24397732 DOI: 10.1586/14737140.2014.867234] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Breast and ovarian cancers are responsible for more than 500,000 female deaths worldwide each year. Fifteen percent of the 230,000 women in the USA diagnosed with breast cancer in 2013 will have triple-negative breast cancer, a disease with few options for treatment, and a twofold greater mortality risk than other breast cancers. The OGF-OGF receptor pathway is present in these cancers, and regulates cell proliferation during homeostasis and disease. OGF is a tonically active peptide that inhibits DNA synthesis by upregulation of cyclin-dependent inhibitory kinases, without disrupting cell migration, differentiation or apoptosis. OGF receptor is a determinant in the proliferation of triple-negative breast cancer and ovarian cancer, and can be genetically modified to alter neoplastic cell replication in vitro and in nude mice. Preclinical studies warrant the use of OGF alone, or in combination, for treatment of triple-negative breast and ovarian cancer patients.
Collapse
Affiliation(s)
- Patricia J McLaughlin
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | |
Collapse
|
30
|
Ebrahimpour S, Tabari MA, Youssefi MR, Aghajanzadeh H, Behzadi MY. Synergistic effect of aged garlic extract and naltrexone on improving immune responses to experimentally induced fibrosarcoma tumor in BALB/c mice. Pharmacognosy Res 2013; 5:189-94. [PMID: 23901215 PMCID: PMC3719261 DOI: 10.4103/0974-8490.112426] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 02/11/2013] [Accepted: 05/22/2013] [Indexed: 11/27/2022] Open
Abstract
Background: Garlic, a medicinal plant, and Naltrexone (NTX), an opioid receptor antagonist, both have immunomodulatory and antitumor effects. Current study was designed to evaluate synergistic antitumor effects of aged garlic extract (AGE) and NTX. Materials and Methods: WEHI-164 fibrosarcoma cells were implanted subcutaneously on day 0 into right flank of 80 BALB/c mice at age of 8 weeks. Mice were randomly categorized in four separate groups: The first group received AGE (100 mg/kg, i.p.), the second group received NTX (0.5 mg/kg, i.p.), the third group received both of them, and the fourth group received phosphate buffered saline as control group. Treatments were administered three times per week. Tumor growth was measured and morbidity was recorded. Subpopulations of CD4+/CD8+ T cells were determined using flowcytometery. WEHI-164 cell specific cytotoxicity of splenocytes and in vitro production of interferon-gamma (IFN-γ) and interleukin-4 (IL-4) cytokines were measured. All statistical analyses were conducted with SPSS 16 software and P < 0.05 was considered to be statistically significant. Results: The mice who received AGE+NTX had significantly longer survival time compared with the mice treated with AGE or NTX alone. An enhanced inhibitory effect on tumor growth was seen in combination therapy group. The CD4+/CD8+ ratio and in vitro IFN-γ production of splenocytes were significantly increased in AGE+NTX and NTX groups. WEHI-164 specific cytotoxicity of splenocytes was also significantly increased at 25:1 E:T ratio in AGE+NTX treated mice. Coadministration of AGE with NTX resulted in improvement of immune responses against experimentally implanted fibrosarcoma tumors in BALB/c mice. Conclusions: AGE showed synergistic effects with NTX on inhibition of tumor growth and increment of survival times.
Collapse
Affiliation(s)
- Soheil Ebrahimpour
- Infection Disease and Tropical Medicine Research Center, Babol University of Medical Sciences, Babol, Iran
| | | | | | | | | |
Collapse
|
31
|
Zagon IS, Donahue R, McLaughlin PJ. Targeting the opioid growth factor: Opioid growth factor receptor axis for treatment of human ovarian cancer. Exp Biol Med (Maywood) 2013; 238:579-87. [DOI: 10.1177/1535370213488483] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The opioid growth factor (OGF) – opioid growth factor receptor (OGFr) axis is a biological pathway that is present in human ovarian cancer cells and tissues. OGF, chemically termed [Met5]-enkephalin, is an endogenous opioid peptide that interfaces with OGFr to delay cells moving through the cell cycle by upregulation of cyclin-dependent inhibitory kinase pathways. OGF inhibitory activity is dose dependent, receptor mediated, reversible, protein and RNA dependent, but not related to apoptosis or necrosis. The OGF-OGFr axis can be targeted for treatment of human ovarian cancer by (i) administration of exogenous OGF, (ii) genetic manipulation to over-express OGFr and (iii) use of low dosages of naltrexone, an opioid antagonist, which stimulates production of OGF and OGFr for subsequent interaction following blockade of the receptor. The OGF-OGFr axis may be a feasible target for treatment of cancer of the ovary (i) in a prophylactic fashion, (ii) following cytoreduction or (iii) in conjunction with standard chemotherapy for additive effectiveness. In summary, preclinical data support the transition of these novel therapies for treatment of human ovarian cancer from the bench to bedside to provide additional targets for treatment of this devastating disease.
Collapse
Affiliation(s)
- Ian S Zagon
- Department of Neural and Behavioral Science, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Renee Donahue
- Department of Neural and Behavioral Science, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Patricia J McLaughlin
- Department of Neural and Behavioral Science, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
32
|
Opioid growth factor (OGF) for hepatoblastoma: a novel non-toxic treatment. Invest New Drugs 2012; 31:1066-70. [DOI: 10.1007/s10637-012-9918-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 12/17/2012] [Indexed: 10/27/2022]
|
33
|
Abstract
This paper is the thirty-fourth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2011 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
34
|
Kakar SS, Jala VR, Fong MY. Synergistic cytotoxic action of cisplatin and withaferin A on ovarian cancer cell lines. Biochem Biophys Res Commun 2012; 423:819-25. [PMID: 22713472 DOI: 10.1016/j.bbrc.2012.06.047] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 06/09/2012] [Indexed: 11/18/2022]
Abstract
Cisplatin derivatives are used as the mainline treatment of ovarian cancer, despite their severe side effects and development of resistance. We developed a novel combination therapy by combining cisplatin with withaferin A. Treatment of ovarian cancer cell lines with combination therapy acted synergistically to induce cell death, thus required a lower dose of cisplatin to achieve the same therapeutic effect. WFA and cisplatin combination induced cell death through the generation of reactive oxygen species (ROS) for WFA, while DNA damage for cisplatin, suggesting that cisplatin binds directly to DNA to form adducts while WFA indirectly damages DNA through ROS generation. Our results for the first time suggest that combining low dose of cisplatin with suboptimal dose of WFA can serve as a potential combination therapy for the treatment of ovarian cancer with the potential to minimize/eliminate the side effects associated with high doses of cisplatin.
Collapse
Affiliation(s)
- Sham S Kakar
- Department of Physiology and Biophysics, University of Louisville, Louisville, KY 40202, United States.
| | | | | |
Collapse
|
35
|
Donahue RN, McLaughlin PJ, Zagon IS. Under-expression of the opioid growth factor receptor promotes progression of human ovarian cancer. Exp Biol Med (Maywood) 2012; 237:167-77. [DOI: 10.1258/ebm.2011.011321] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The opioid growth factor (OGF) and its receptor, OGFr, serve as a tonically active inhibitory axis regulating the proliferation of human ovarian cancer cells. In the present study, we have investigated the repercussion on the progression of this deadly neoplasia when cells are engineered to molecularly under-express OGFr. shRNA constructs were used to knockdown OGFr in SKOV-3 cells; two clonal cell lines were examined. OGFr protein expression was decreased up to 73% in clones compared with wild-type (WT) and empty vector (EV) controls. OGFr-binding assays of clones revealed 50–55% decreases in binding capacity compared with control cells; binding affinity was comparable in all groups. Cell number in clones was increased 33–132%, and doubling times decreased 29–35%, compared with WT and EV cultures. Addition of exogenous OGF or naltrexone did not affect cell number in cultures with silenced OGFr. DNA synthesis of clonal cell lines was increased 136–146% from the WT and EV groups; no changes were noted in cell survival. Nude mice injected subcutaneously with cells under-expressing OGFr had an increased tumor incidence, decreased latency to tumor formation, increased tumor volume and decreased OGFr expression in tumors compared with WT and EV controls. OGF treatment in mice with WT or EV tumors, but not OGFr under-expressing tumors, inhibited tumor volume and weight. Collectively, these data demonstrate the critical nature of the OGF–OGFr axis as a determinant of the progression of human ovarian cancer, and suggest that attenuation of this system has an important bearing on the survival of these patients.
Collapse
Affiliation(s)
- Renee N Donahue
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, College of Medicine, Hershey, PA 17033–0850, USA
| | - Patricia J McLaughlin
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, College of Medicine, Hershey, PA 17033–0850, USA
| | - Ian S Zagon
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, College of Medicine, Hershey, PA 17033–0850, USA
| |
Collapse
|
36
|
Fanning J, Hossler CA, Kesterson JP, Donahue RN, McLaughlin PJ, Zagon IS. Expression of the opioid growth factor–opioid growth factor receptor axis in human ovarian cancer. Gynecol Oncol 2012; 124:319-24. [DOI: 10.1016/j.ygyno.2011.10.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 10/19/2011] [Accepted: 10/24/2011] [Indexed: 12/30/2022]
|
37
|
Influence of morphine on pericyte-endothelial interaction: implications for antiangiogenic therapy. JOURNAL OF ONCOLOGY 2012; 2012:458385. [PMID: 22315595 PMCID: PMC3270445 DOI: 10.1155/2012/458385] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 10/07/2011] [Indexed: 12/12/2022]
Abstract
Morphine stimulates tumor angiogenesis and cancer progression in mice. We examined if morphine influences endothelial-pericyte interaction via platelet-derived growth factor-BB (PDGF-BB) and PDGF receptor-β (PDGFR-β). Clinically relevant doses of morphine stimulated PDGF-BB secretion from human umbilical vein endothelial cells and activated PDGFR-β and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) phosphorylation in human pericytes. These in vitro effects of morphine were translated into promotion of tumor angiogenesis in a transgenic mice model of breast cancer when treated with clinically used dose of morphine. Increased vessel-associated immunoreactivity of desmin and PDGFR-β was observed on pericytes in tumors of morphine-treated mice. These data suggest that morphine potentiates endothelial-pericyte interaction via PDGF-BB/PDGFR-β signaling and promotes tumor angiogenesis, pericyte recruitment, and coverage of tumor vessels. We speculate that morphine may impair the effectiveness of antiangiogenic therapy by influencing vascular pericyte coverage.
Collapse
|