1
|
Jin X, Han H, Liang Q. Effects of surgical trauma and intraoperative blood loss on tumour progression. Front Oncol 2024; 14:1412367. [PMID: 38912060 PMCID: PMC11190163 DOI: 10.3389/fonc.2024.1412367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 05/22/2024] [Indexed: 06/25/2024] Open
Abstract
Surgery is the primary treatment of choice for tumours, and improves prognosis, prolongs survival and is potentially curative. Previous studies have described the effects of anaesthesia and changes in the neuroendocrine, circulatory and sympathetic nervous systems on postoperative cancer progression. There is growing evidence that intraoperative blood loss is an independent prognostic factor for tumour recurrence, postoperative inflammation is a predictor of cancer prognosis, and immunosuppressive status correlates with the degree of surgical damage. This paper outlines the potential mechanisms by which blood loss, surgical trauma and postoperative immunosuppressive status contribute to tumour growth and recurrence by reducing intraoperative haemorrhage and perioperative immunotherapy, thereby reducing tumour growth and recurrence, and improving long-term prognosis.
Collapse
Affiliation(s)
| | | | - Qilian Liang
- Oncology Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
2
|
Zhang D, Jiang J, Liu J, Zhu T, Huang H, Zhou C. Effects of Perioperative Epidural Analgesia on Cancer Recurrence and Survival. Front Oncol 2022; 11:798435. [PMID: 35071003 PMCID: PMC8766638 DOI: 10.3389/fonc.2021.798435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/10/2021] [Indexed: 02/05/2023] Open
Abstract
Surgical resection is the main curative avenue for various cancers. Unfortunately, cancer recurrence following surgery is commonly seen, and typically results in refractory disease and death. Currently, there is no consensus whether perioperative epidural analgesia (EA), including intraoperative and postoperative epidural analgesia, is beneficial or harmful on cancer recurrence and survival. Although controversial, mounting evidence from both clinical and animal studies have reported perioperative EA can improve cancer recurrence and survival via many aspects, including modulating the immune/inflammation response and reducing the use of anesthetic agents like inhalation anesthetics and opioids, which are independent risk factors for cancer recurrence. However, these results depend on the cancer types, cancer staging, patients age, opioids use, and the duration of follow-up. This review will summarize the effects of perioperative EA on the oncological outcomes of patients after cancer surgery.
Collapse
Affiliation(s)
- Donghang Zhang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China.,Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Jingyao Jiang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China.,Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Han Huang
- Department of Anesthesiology & Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital of Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Carney CP, Pandey N, Kapur A, Woodworth GF, Winkles JA, Kim AJ. Harnessing nanomedicine for enhanced immunotherapy for breast cancer brain metastases. Drug Deliv Transl Res 2021; 11:2344-2370. [PMID: 34716900 PMCID: PMC8568876 DOI: 10.1007/s13346-021-01039-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2021] [Indexed: 12/15/2022]
Abstract
Brain metastases (BMs) are the most common type of brain tumor, and the incidence among breast cancer (BC) patients has been steadily increasing over the past two decades. Indeed, ~ 30% of all patients with metastatic BC will develop BMs, and due to few effective treatments, many will succumb to the disease within a year. Historically, patients with BMs have been largely excluded from clinical trials investigating systemic therapies including immunotherapies (ITs) due to limited brain penetration of systemically administered drugs combined with previous assumptions that BMs are poorly immunogenic. It is now understood that the central nervous system (CNS) is an immunologically distinct site and there is increasing evidence that enhancing immune responses to BCBMs will improve patient outcomes and the efficacy of current treatment regimens. Progress in IT for BCBMs, however, has been slow due to several intrinsic limitations to drug delivery within the brain, substantial safety concerns, and few known targets for BCBM IT. Emerging studies demonstrate that nanomedicine may be a powerful approach to overcome such limitations, and has the potential to greatly improve IT strategies for BMs specifically. This review summarizes the evidence for IT as an effective strategy for BCBM treatment and focuses on the nanotherapeutic strategies currently being explored for BCBMs including targeting the blood-brain/tumor barrier (BBB/BTB), tumor cells, and tumor-supporting immune cells for concentrated drug release within BCBMs, as well as use of nanoparticles (NPs) for delivering immunomodulatory agents, for inducing immunogenic cell death, or for potentiating anti-tumor T cell responses.
Collapse
Affiliation(s)
- Christine P Carney
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Nikhil Pandey
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Anshika Kapur
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jeffrey A Winkles
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Surgery and Neurosurgery, University of Maryland School of Medicine, 800 West Baltimore St., Baltimore, MD, 21201, USA.
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA.
- Departments of Neurosurgery, Pharmacology, and Pharmaceutical Sciences, University of Maryland School of Medicine, 655 W Baltimore St., Baltimore, MD, 21201, USA.
| |
Collapse
|
4
|
Surgical Stress Promotes Tumor Progression: A Focus on the Impact of the Immune Response. J Clin Med 2020; 9:jcm9124096. [PMID: 33353113 PMCID: PMC7766515 DOI: 10.3390/jcm9124096] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/16/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022] Open
Abstract
Despite advances in systemic therapies, surgery is crucial for the management of solid malignancy. There is increasing evidence suggesting that the body's response to surgical stress resulting from tumor resection has direct effects on tumor cells or can alter the tumor microenvironment. Surgery can lead to the activation of early and key components of the innate and adaptative immune systems. Platelet activation and the subsequent pro-coagulation state can accelerate the growth of micrometastases. Neutrophil extracellular traps (NETs), an extracellular network of DNA released by neutrophils in response to inflammation, promote the adhesion of circulating tumor cells and the growth of existing micrometastatic disease. In addition, the immune response following cancer surgery can modulate the tumor immune microenvironment by promoting an immunosuppressive state leading to impaired recruitment of natural killer (NK) cells and regulatory T cells (Tregs). In this review, we will summarize the current understanding of mechanisms of tumor progression secondary to surgical stress. Furthermore, we will describe emerging and novel peri-operative solutions to decrease pro-tumorigenic effects from surgery.
Collapse
|
5
|
Seiler A, Sood AK, Jenewein J, Fagundes CP. Can stress promote the pathophysiology of brain metastases? A critical review of biobehavioral mechanisms. Brain Behav Immun 2020; 87:860-880. [PMID: 31881262 DOI: 10.1016/j.bbi.2019.12.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 12/15/2019] [Accepted: 12/20/2019] [Indexed: 01/20/2023] Open
Abstract
Chronic stress can promote tumor growth and progression through immunosuppressive effects and bi-directional interactions between tumor cells and their microenvironment. β-Adrenergic receptor signaling plays a critical role in mediating stress-related effects on tumor progression. Stress-related mechanisms that modulate the dissemination of tumor cells to the brain have received scant attention. Brain metastases are highly resistant to chemotherapy and contribute considerably to morbidity and mortality in various cancers, occurring in up to 20% of patients in some cancer types. Understanding the mechanisms promoting brain metastasis could help to identify interventions that improve disease outcomes. In this review, we discuss biobehavioral, sympathetic, neuroendocrine, and immunological mechanisms by which chronic stress can impact tumor progression and metastatic dissemination to the brain. The critical role of the inflammatory tumor microenvironment in tumor progression and metastatic dissemination to the brain, and its association with stress pathways are delineated. We also discuss translational implications for biobehavioral and pharmacological interventions.
Collapse
Affiliation(s)
- Annina Seiler
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland.
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Josef Jenewein
- Clinic Zugersee, Center for Psychiatry and Psychotherapy, Oberwil-Zug, Switzerland
| | - Christopher P Fagundes
- Department of Psychology, Rice University, Houston, TX, United States; Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
6
|
Prevention of liver metastases through perioperative acute CpG-C immune stimulation. Cancer Immunol Immunother 2020; 69:2021-2031. [PMID: 32405793 DOI: 10.1007/s00262-020-02596-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 04/27/2020] [Indexed: 12/21/2022]
Abstract
Following excision of colorectal tumors, metastatic disease is prevalent, primarily occurs in the liver, and is highly predictive of poor prognosis. The perioperative period is now recognized as critical in determining the incidence of postoperative metastases and long-term cancer outcomes. Thus, various perioperative prophylactic interventions are currently studied during this time frame. However, immune stimulation during the perioperative period has rarely been attempted due to specific contraindications to surgery and various adverse effects. Here, to prevent liver metastases, we perioperatively employed a TLR-9 agonist, CpG-C, which exhibits minimal pyrogenic and other adverse effects in patients. We found that marginating-hepatic (MH) cells in BALB/c mice contained high percentage of NK cells, but exhibited negligible NK cytotoxicity, as previously reported in humans. However, a single CpG-C administration (25-100 µg/mouse) doubled MH-NK cell numbers, increased NK cell activation and maturation markers (NKp46, CD11b), decreased the inhibitory NKG2A ligand, and dramatically increased MH-NK-cell cytotoxicity against the syngeneic CT26 colon cancer line. Moreover, in operated mice, this innocuous intervention also markedly improved resistance to CT26 and MC38 hepatic metastases in BALB/c and C57BL/6 mice, respectively. Beneficial effects of CpG-C were mediated through activation of MH-NK cells, as indicated by an in vivo NK depletion study. Last, CpG-C protected against surgery-induced suppression of MH-NK cytotoxicity and improved their activation indices. Thus, we suggest that systemic perioperative CpG-C treatment should be considered and studied as a novel therapeutic approach to improve long-term cancer outcomes in colorectal cancer patients.
Collapse
|
7
|
Chen Z, Zhang P, Xu Y, Yan J, Liu Z, Lau WB, Lau B, Li Y, Zhao X, Wei Y, Zhou S. Surgical stress and cancer progression: the twisted tango. Mol Cancer 2019; 18:132. [PMID: 31477121 PMCID: PMC6717988 DOI: 10.1186/s12943-019-1058-3] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022] Open
Abstract
Surgical resection is an important avenue for cancer treatment, which, in most cases, can effectively alleviate the patient symptoms. However, accumulating evidence has documented that surgical resection potentially enhances metastatic seeding of tumor cells. In this review, we revisit the literature on surgical stress, and outline the mechanisms by which surgical stress, including ischemia/reperfusion injury, activation of sympathetic nervous system, inflammation, systemically hypercoagulable state, immune suppression and effects of anesthetic agents, promotes tumor metastasis. We also propose preventive strategies or resolution of tumor metastasis caused by surgical stress.
Collapse
Affiliation(s)
- Zhiwei Chen
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China
| | - Peidong Zhang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China
| | - Ya Xu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China.,Deyang People's Hospital, Deyang, Sichuan, People's Republic of China
| | - Jiahui Yan
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China
| | - Zixuan Liu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University Hospital, Philadelphia, USA
| | - Bonnie Lau
- Department of Surgery, Emergency Medicine, Kaiser Santa Clara Medical Center, Affiliate of Stanford University, Stanford, USA
| | - Ying Li
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong, People's Republic of China
| | - Xia Zhao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China
| | - Yuquan Wei
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
8
|
Matzner P, Sorski L, Haldar R, Shaashua L, Benbenishty A, Lavon H, Azan Y, Sandbank E, Melamed R, Rosenne E, Ben-Eliyahu S. Deleterious synergistic effects of distress and surgery on cancer metastasis: Abolishment through an integrated perioperative immune-stimulating stress-inflammatory-reducing intervention. Brain Behav Immun 2019; 80:170-178. [PMID: 30851377 DOI: 10.1016/j.bbi.2019.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 02/26/2019] [Accepted: 03/05/2019] [Indexed: 01/21/2023] Open
Abstract
The perioperative period holds disproportionate impact on long-term cancer outcomes. Nevertheless, perioperative interventions to improve long-term cancer outcomes are not clinical routines, including perioperative stress-reducing or immune-stimulating approaches. Here, mimicking the clinical setting of pre-operative distress, followed by surgery, we examined the separate and combined effects of these events on the efficacy of pre-operative immune stimulation in rats and mice, and on post-operative resistance to tumor metastasis of the syngeneic mammary adenocarcinoma MADB106 in F344 rats and the CT26 colon carcinoma in Balb/C mice. The novel immune stimulating agents, GLA-SE or CpG-C (TLR-4 and TLR-9 agonists, respectively), were employed pre-operatively. Sixteen hours of pre-operative behavioral stressors (i) lowered CpG-C induced plasma IL-12 levels, and reduced resistance to MADB106 and CT-26 experimental metastases, and (ii) worsened the deleterious effects of laparotomy on metastasis in both tumor models. In rats, these effects of pre-operative stress were further studied and successfully abolished by the glucocorticoid receptor antagonist RU-486. Additionally, in vitro studies indicated the dampening effect of corticosterone on immune stimulation. Last, we tested a perioperative integrated intervention in the context of pre-operative stress and laparotomy, based on (i) antagonizing the impact of glucocorticoids before surgery, (ii) activating anti-metastatic immunity perioperatively, and (iii) blocking excessive operative and post-operative adrenergic and prostanoid responses. This integrated intervention successfully and completely abolished the deleterious effects of stress and of surgery on post-operative resistance to experimental metastasis. Such and similar integrated approaches can be studied clinically in cancer patients.
Collapse
Affiliation(s)
- Pini Matzner
- Neuroimmunology Research Unit, Sagol School of Neuroscience, and The School of Psychological Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Liat Sorski
- Neuroimmunology Research Unit, Sagol School of Neuroscience, and The School of Psychological Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Rita Haldar
- Neuroimmunology Research Unit, Sagol School of Neuroscience, and The School of Psychological Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Lee Shaashua
- Neuroimmunology Research Unit, Sagol School of Neuroscience, and The School of Psychological Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Amit Benbenishty
- Neuroimmunology Research Unit, Sagol School of Neuroscience, and The School of Psychological Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Hagar Lavon
- Neuroimmunology Research Unit, Sagol School of Neuroscience, and The School of Psychological Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Yosi Azan
- Neuroimmunology Research Unit, Sagol School of Neuroscience, and The School of Psychological Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Elad Sandbank
- Neuroimmunology Research Unit, Sagol School of Neuroscience, and The School of Psychological Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Rivka Melamed
- Neuroimmunology Research Unit, Sagol School of Neuroscience, and The School of Psychological Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Ella Rosenne
- Neuroimmunology Research Unit, Sagol School of Neuroscience, and The School of Psychological Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Shamgar Ben-Eliyahu
- Neuroimmunology Research Unit, Sagol School of Neuroscience, and The School of Psychological Sciences, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
9
|
Benbenishty A, Gadrich M, Cottarelli A, Lubart A, Kain D, Amer M, Shaashua L, Glasner A, Erez N, Agalliu D, Mayo L, Ben-Eliyahu S, Blinder P. Prophylactic TLR9 stimulation reduces brain metastasis through microglia activation. PLoS Biol 2019; 17:e2006859. [PMID: 30921319 PMCID: PMC6469801 DOI: 10.1371/journal.pbio.2006859] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 04/17/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023] Open
Abstract
Brain metastases are prevalent in various types of cancer and are often terminal, given the low efficacy of available therapies. Therefore, preventing them is of utmost clinical relevance, and prophylactic treatments are perhaps the most efficient strategy. Here, we show that systemic prophylactic administration of a toll-like receptor (TLR) 9 agonist, CpG-C, is effective against brain metastases. Acute and chronic systemic administration of CpG-C reduced tumor cell seeding and growth in the brain in three tumor models in mice, including metastasis of human and mouse lung cancer, and spontaneous melanoma-derived brain metastasis. Studying mechanisms underlying the therapeutic effects of CpG-C, we found that in the brain, unlike in the periphery, natural killer (NK) cells and monocytes are not involved in controlling metastasis. Next, we demonstrated that the systemically administered CpG-C is taken up by endothelial cells, astrocytes, and microglia, without affecting blood-brain barrier (BBB) integrity and tumor brain extravasation. In vitro assays pointed to microglia, but not astrocytes, as mediators of CpG- C effects through increased tumor killing and phagocytosis, mediated by direct microglia-tumor contact. In vivo, CpG-C-activated microglia displayed elevated mRNA expression levels of apoptosis-inducing and phagocytosis-related genes. Intravital imaging showed that CpG-C-activated microglia cells contact, kill, and phagocytize tumor cells in the early stages of tumor brain invasion more than nonactivated microglia. Blocking in vivo activation of microglia with minocycline, and depletion of microglia with a colony-stimulating factor 1 inhibitor, indicated that microglia mediate the antitumor effects of CpG-C. Overall, the results suggest prophylactic CpG-C treatment as a new intervention against brain metastasis, through an essential activation of microglia.
Collapse
Affiliation(s)
- Amit Benbenishty
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
- Neurobiology Department, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Meital Gadrich
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- School for Molecular Cell Biology & Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Azzurra Cottarelli
- Department of Neurology, Columbia University Medical Center, New York, New York, United States of America
| | - Alisa Lubart
- Neurobiology Department, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - David Kain
- Neurobiology Department, Tel Aviv University, Tel Aviv, Israel
| | - Malak Amer
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lee Shaashua
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ariella Glasner
- The Lautenberg Centre for General and Tumor Immunology, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dritan Agalliu
- Department of Neurology, Columbia University Medical Center, New York, New York, United States of America
| | - Lior Mayo
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- School for Molecular Cell Biology & Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Shamgar Ben-Eliyahu
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Pablo Blinder
- Neurobiology Department, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
10
|
Bakos O, Lawson C, Rouleau S, Tai LH. Combining surgery and immunotherapy: turning an immunosuppressive effect into a therapeutic opportunity. J Immunother Cancer 2018; 6:86. [PMID: 30176921 PMCID: PMC6122574 DOI: 10.1186/s40425-018-0398-7] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 08/16/2018] [Indexed: 12/27/2022] Open
Abstract
Background Cancer surgery is necessary and life-saving. However, the majority of patients develop postoperative recurrence and metastasis, which are the main causes of cancer-related deaths. The postoperative stress response encompasses a broad set of physiological changes that have evolved to safeguard the host following major tissue trauma. These stress responses, however, intersect with cellular mediators and signaling pathways that contribute to cancer proliferation. Main Previous descriptive and emerging mechanistic studies suggest that the surgery-induced prometastatic effect is linked to impairment of both innate and adaptive immunity. Existing studies that combine surgery and immunotherapies have revealed that this combination strategy is not straightforward and patients have experienced both therapeutic benefit and drawbacks. This review will specifically assess the immunological pathways that are disrupted by oncologic surgical stress and provide suggestions for rationally combining cancer surgery with immunotherapies to improve immune and treatment outcomes. Short conclusion Given the prevalence of surgery as frontline therapy for solid cancers, the emerging data on postoperative immunosuppression and the rapid development of immunotherapy for oncologic treatment, we believe that future targeted studies of perioperative immunotherapy are warranted.
Collapse
Affiliation(s)
- Orneala Bakos
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Christine Lawson
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Samuel Rouleau
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Lee-Hwa Tai
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada. .,Centre de Recherche Clinique de Centre Hospitalier de l'Université de Sherbrooke (CHUS), Room 4853, 3001, 12e Avenue Nord, Sherbrooke, Québec, J1H 5N4, Canada.
| |
Collapse
|
11
|
Alvarado A, Faustino-Rocha AI, Colaço B, Oliveira PA. Experimental mammary carcinogenesis - Rat models. Life Sci 2017; 173:116-134. [PMID: 28188729 DOI: 10.1016/j.lfs.2017.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/26/2017] [Accepted: 02/06/2017] [Indexed: 12/22/2022]
Abstract
Mammary cancer is one of the most common cancers, victimizing more than half a million of women worldwide every year. Despite all the studies in this field, the current therapeutic approaches are not effective and have several devastating effects for patients. In this way, the need to better understand the mammary cancer biopathology and find effective therapies led to the development of several rodent models over years. With this review, the authors intended to provide the readers with an overview of the rat models used to study mammary carcinogenesis, with a special emphasis on chemically-induced models.
Collapse
Affiliation(s)
- Antonieta Alvarado
- Área de Patología, Decanato de Ciencias Veterinarias, Universidad Centroccidental "Lisandro Alvarado", UCLA, Lara, Venezuela; Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Ana I Faustino-Rocha
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, UTAD, Vila Real, Portugal
| | - Bruno Colaço
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Department of Zootechnics, School of Agrarian and Veterinary Sciences, UTAD, Vila Real, Portugal
| | - Paula A Oliveira
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, UTAD, Vila Real, Portugal.
| |
Collapse
|
12
|
Stress impairs the efficacy of immune stimulation by CpG-C: Potential neuroendocrine mediating mechanisms and significance to tumor metastasis and the perioperative period. Brain Behav Immun 2016; 56:209-220. [PMID: 26944000 PMCID: PMC4917466 DOI: 10.1016/j.bbi.2016.02.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 02/28/2016] [Accepted: 02/29/2016] [Indexed: 12/29/2022] Open
Abstract
We recently reported that immune stimulation can be compromised if animals are simultaneously subjected to stressful conditions. To test the generalizability of these findings, and to elucidate neuroendocrine mediating mechanisms, we herein employed CpG-C, a novel TLR-9 immune-stimulating agent. Animals were subjected to ongoing stress (20-h of wet cage exposure) during CpG-C treatment, and antagonists to glucocorticoids, β-adrenoceptor, COX2, or opioids were employed (RU486, nadolol, etodolac, naltrexone). In F344 rats, marginating-pulmonary NK cell numbers and cytotoxicity were studied, and the NK-sensitive MADB106 experimental metastasis model was used. In Balb/C mice, experimental hepatic metastases of the CT-26 colon tumor were studied; and in C57BL/6J mice, survival rates following excision of B16 melanoma was assessed - both mouse tumor models involved surgical stress. The findings indicated that simultaneous blockade of glucocorticoid and β-adrenergic receptors improved CpG-C efficacy against MADB106 metastasis. In mice bearing B16 melanoma, long-term survival rate was improved by CpG-C only when employed simultaneously with blockers of glucocorticoids, catecholamines, and prostaglandins. Prolonged stress impaired CpG-C efficacy in potentiating NK activity, and in resisting MADB106 metastasis in both sexes, as also supported by in vitro studies. This latter effect was not blocked by any of the antagonists or by adrenalectomy. In the CT26 model, prolonged stress only partially reduced the efficacy of CpG-C. Overall, our findings indicate that ongoing behavioral stress and surgery can jeopardize immune-stimulatory interventions and abolish their beneficial metastasis-reducing impacts. These findings have implications for the clinical setting, which often involve psychological and physiological stress responses during immune-stimulation.
Collapse
|
13
|
Matzner P, Sorski L, Shaashua L, Elbaz E, Lavon H, Melamed R, Rosenne E, Gotlieb N, Benbenishty A, Reed SG, Ben-Eliyahu S. Perioperative treatment with the new synthetic TLR-4 agonist GLA-SE reduces cancer metastasis without adverse effects. Int J Cancer 2015; 138:1754-64. [PMID: 26453448 DOI: 10.1002/ijc.29885] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 09/28/2015] [Indexed: 01/08/2023]
Abstract
The use of TLR agonists as an anti-cancer treatment is gaining momentum given their capacity to activate various host cellular responses through the secretion of inflammatory cytokines and type-I interferons. It is now also recognized that the perioperative period is a window of opportunity for various interventions aiming at reducing the risk of cancer metastases-the major cause of cancer related death. However, immune-stimulatory approach has not been used perioperatively given several contraindications to surgery. To overcome these obstacles, in this study, we used the newly introduced, fully synthetic TLR-4 agonist, Glucopyranosyl Lipid-A (GLA-SE), in various models of cancer metastases, and in the context of acute stress or surgery. Without exerting evident adverse effects, a single systemic administration of GLA-SE rapidly and dose dependently elevated both innate and adaptive immunity in the circulation, lungs and the lymphatic system. Importantly, GLA-SE treatment led to reduced metastatic development of a mammary adenocarcinoma and a colon carcinoma by approximately 40-75% in F344 rats and BALB/c mice, respectively, at least partly through elevating marginating-pulmonary NK cell cytotoxicity. GLA-SE is safe and well tolerated in humans, and currently is used as an adjuvant in phase-II clinical trials. Given that the TLR-4 receptor and its signaling cascade is highly conserved throughout evolution, our current results suggest that GLA-SE may be a promising immune stimulatory agent in the context of oncological surgeries, aiming to reduce long-term cancer recurrence.
Collapse
Affiliation(s)
- Pini Matzner
- Neuro-Immunology Research Unit, School of Psychological Sciences Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Liat Sorski
- Neuro-Immunology Research Unit, School of Psychological Sciences Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Lee Shaashua
- Neuro-Immunology Research Unit, School of Psychological Sciences Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Ely Elbaz
- Neuro-Immunology Research Unit, School of Psychological Sciences Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Hagar Lavon
- Neuro-Immunology Research Unit, School of Psychological Sciences Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Rivka Melamed
- Neuro-Immunology Research Unit, School of Psychological Sciences Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Ella Rosenne
- Neuro-Immunology Research Unit, School of Psychological Sciences Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Neta Gotlieb
- Department of Psychology, University of California, Berkely, California
| | - Amit Benbenishty
- Neuro-Immunology Research Unit, School of Psychological Sciences Tel-Aviv University, Tel Aviv-Yafo, Israel.,Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Steve G Reed
- Infectious Disease Research Institute (IDRI), Seattle, Washington
| | - Shamgar Ben-Eliyahu
- Neuro-Immunology Research Unit, School of Psychological Sciences Tel-Aviv University, Tel Aviv-Yafo, Israel.,Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv-Yafo, Israel
| |
Collapse
|
14
|
Scanzano A, Cosentino M. Adrenergic regulation of innate immunity: a review. Front Pharmacol 2015; 6:171. [PMID: 26321956 PMCID: PMC4534859 DOI: 10.3389/fphar.2015.00171] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/31/2015] [Indexed: 12/24/2022] Open
Abstract
The sympathetic nervous system has a major role in the brain-immune cross-talk, but few information exist on the sympathoadrenergic regulation of innate immune system. The aim of this review is to summarize available knowledge regarding the sympathetic modulation of the innate immune response, providing a rational background for the possible repurposing of adrenergic drugs as immunomodulating agents. The cells of immune system express adrenoceptors (AR), which represent the target for noradrenaline and adrenaline. In human neutrophils, adrenaline and noradrenaline inhibit migration, CD11b/CD18 expression, and oxidative metabolism, possibly through β-AR, although the role of α1- and α2-AR requires further investigation. Natural Killer express β-AR, which are usually inhibitory. Monocytes express β-AR and their activation is usually antiinflammatory. On murine Dentritic cells (DC), β-AR mediate sympathetic influence on DC-T cells interactions. In human DC β2-AR may affect Th1/2 differentiation of CD4+ T cells. In microglia and in astrocytes, β2-AR dysregulation may contribute to neuroinflammation in autoimmune and neurodegenerative disease. In conclusion, extensive evidence supports a critical role for adrenergic mechanisms in the regulation of innate immunity, in peripheral tissues as well as in the CNS. Sympathoadrenergic pathways in the innate immune system may represent novel antiinflammatory and immunomodulating targets with significant therapeutic potential.
Collapse
Affiliation(s)
- Angela Scanzano
- Center for Research in Medical Pharmacology, University of Insubria Varese, Italy
| | - Marco Cosentino
- Center for Research in Medical Pharmacology, University of Insubria Varese, Italy
| |
Collapse
|
15
|
Kadosawa T, Watabe A. The effects of surgery-induced immunosuppression and angiogenesis on tumour growth. Vet J 2015; 205:175-9. [DOI: 10.1016/j.tvjl.2015.04.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 03/19/2015] [Accepted: 04/07/2015] [Indexed: 01/26/2023]
|
16
|
De Lorenzo BHP, de Oliveira Marchioro L, Greco CR, Suchecki D. Sleep-deprivation reduces NK cell number and function mediated by β-adrenergic signalling. Psychoneuroendocrinology 2015; 57:134-43. [PMID: 25929826 DOI: 10.1016/j.psyneuen.2015.04.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 04/06/2015] [Accepted: 04/06/2015] [Indexed: 11/28/2022]
Abstract
Reduction of sleep time triggers a stress response, leading to augmented levels of glucocorticoids and adrenaline. These hormones regulate components of the innate immune system such as natural killer (NK) and NKT cells. In the present study, we sought to investigate whether and how stress hormones could alter the population and function of NK and NKT cells of mice submitted to different lengths of paradoxical sleep deprivation (PSD, from 24 to 72 h). Results showed that 72h of PSD decreased not only NK and NKT cell counts, but also their cytotoxic activity against B16F10 melanoma cells in vitro. Propranolol treatment during PSD reversed these effects, indicating a major inhibitory role of beta-adrenergic receptors (β-AR) on NK cells function. Moreover, both corticosterone plasma levels and expression of beta 2-adrenergic receptors (β2-AR) in NK cells increased by 48 h of PSD. In vitro incubation of NK cells with dexamethasone augmented the level of β2-AR in the cell surface, suggesting that glucocorticoids could induce β2-AR expression. In summary, we propose that reduction of NK and NKT cell number and cytotoxic activity appears to be mediated by glucocorticoids-induced increased expression of β2-AR in these cells.
Collapse
Affiliation(s)
- Beatriz H P De Lorenzo
- Department of Psychobiology, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925 - 1° andar, Vila Clementino, 04024-002 São Paulo, SP, Brazil; Centro Universitário São Camilo, Avenida Nazaré, 1501, Ipiranga, 04263-200 São Paulo, SP, Brazil.
| | - Laís de Oliveira Marchioro
- Department of Psychobiology, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925 - 1° andar, Vila Clementino, 04024-002 São Paulo, SP, Brazil
| | - Carollina Ribeiro Greco
- Department of Psychobiology, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925 - 1° andar, Vila Clementino, 04024-002 São Paulo, SP, Brazil
| | - Deborah Suchecki
- Department of Psychobiology, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925 - 1° andar, Vila Clementino, 04024-002 São Paulo, SP, Brazil
| |
Collapse
|
17
|
Horowitz M, Neeman E, Sharon E, Ben-Eliyahu S. Exploiting the critical perioperative period to improve long-term cancer outcomes. Nat Rev Clin Oncol 2015; 12:213-26. [PMID: 25601442 PMCID: PMC5497123 DOI: 10.1038/nrclinonc.2014.224] [Citation(s) in RCA: 333] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Evidence suggests that the perioperative period and the excision of the primary tumour can promote the development of metastases—the main cause of cancer-related mortality. This Review first presents the assertion that the perioperative timeframe is pivotal in determining long-term cancer outcomes, disproportionally to its short duration (days to weeks). We then analyse the various aspects of surgery, and their consequent paracrine and neuroendocrine responses, which could facilitate the metastatic process by directly affecting malignant tissues, and/or through indirect pathways, such as immunological perturbations. We address the influences of surgery-related anxiety and stress, nutritional status, anaesthetics and analgesics, hypothermia, blood transfusion, tissue damage, and levels of sex hormones, and point at some as probable deleterious factors. Through understanding these processes and reviewing empirical evidence, we provide suggestions for potential new perioperative approaches and interventions aimed at attenuating deleterious processes and ultimately improving treatment outcomes. Specifically, we highlight excess perioperative release of catecholamines and prostaglandins as key deleterious mediators of surgery, and we recommend blockade of these responses during the perioperative period, as well as other low-risk, low-cost interventions. The measures described in this Review could transform the perioperative timeframe from a prominent facilitator of metastatic progression, to a window of opportunity for arresting and/or eliminating residual disease, potentially improving long-term survival rates in patients with cancer.
Collapse
Affiliation(s)
- Maya Horowitz
- School of Psychological Sciences, Sharet Building, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Elad Neeman
- School of Psychological Sciences, Sharet Building, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Eran Sharon
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petach-Tikva 49100, Israel
| | - Shamgar Ben-Eliyahu
- School of Psychological Sciences, Sharet Building, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
18
|
Rosenne E, Sorski L, Shaashua L, Neeman E, Matzner P, Levi B, Ben-Eliyahu S. In vivo suppression of NK cell cytotoxicity by stress and surgery: glucocorticoids have a minor role compared to catecholamines and prostaglandins. Brain Behav Immun 2014; 37:207-19. [PMID: 24333572 PMCID: PMC4322769 DOI: 10.1016/j.bbi.2013.12.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 11/28/2013] [Accepted: 12/06/2013] [Indexed: 11/30/2022] Open
Abstract
Most in vitro and ex-vivo studies indicate a profound suppression of NK cell cytotoxicity (NKCC) by glucocorticoids; while catecholamines and prostaglandins were reported both to suppress and to enhance NKCC. However, methodological considerations hinder our ability to deduce from these findings to the impact of endogenous release of these factors on in vivo levels of NKCC and their implications to NK-dependent resistance to pathologies in living humans or animals. Here we used an in vivo approach that sensitively and specifically reflects NKCC in living F344 rats, based on lung clearance of NK-sensitive tumor cells (MADB106), and based on comparing effects between NK-intact and NK-depleted rats. To study the role of corticosterone, epinephrine, and prostaglandins, we administered these factors to rats, or antagonized their endogenous release following different stress paradigms or surgery. The results indicated that endogenous or exogenous elevated corticosterone levels can suppress in vivo NKCC levels, but only under some conditions, and mostly secondarily to the NK-suppressing impact of epinephrine. Specifically, corticosterone-induced NKCC suppression occurred (i) only under prolonged, but not short exposure to stress, and mainly in males; (ii) was smaller than the prominent impact of epinephrine; (iii) was mostly ascribed to corticosterone-induced potentiation of the effects of epinephrine or/and prostaglandins; and (iv) was completely abolished through antagonizing epinephrine or/and prostaglandins. Overall, these findings markedly limit the significance of stress/surgery-induced corticosterone release in the in vivo suppression of NKCC, and highlight the blockade of epinephrine or/and prostaglandins as effective and clinically feasible approaches to overcome such immuno-suppressive effects.
Collapse
Affiliation(s)
- Ella Rosenne
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv, Israel
| | - Liat Sorski
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv, Israel
| | - Lee Shaashua
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv, Israel
| | - Elad Neeman
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv, Israel
| | - Pini Matzner
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv, Israel
| | - Ben Levi
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv, Israel
| | - Shamgar Ben-Eliyahu
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
19
|
Neeman E, Ben-Eliyahu S. Surgery and stress promote cancer metastasis: new outlooks on perioperative mediating mechanisms and immune involvement. Brain Behav Immun 2013; 30 Suppl:S32-40. [PMID: 22504092 PMCID: PMC3423506 DOI: 10.1016/j.bbi.2012.03.006] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 03/24/2012] [Indexed: 12/26/2022] Open
Abstract
Surgery for the removal of a primary tumor presents an opportunity to eradicate cancer or arrest its progression, but is also believed to promote the outbreak of pre-existing micrometastases and the initiation of new metastases. These deleterious effects of surgery are mediated through various mechanisms, including psychological and physiological neuroendocrine and paracrine stress responses elicited by surgery. In this review we (i) describe the many risk factors that arise during the perioperative period, acting synergistically to make this short timeframe critical for determining long-term cancer recurrence, (ii) present newly identified potent immunocyte populations that can destroy autologous tumor cells that were traditionally considered immune-resistant, thus invigorating the notion of immune-surveillance against cancer metastasis, (iii) describe in vivo evidence in cancer patients that support a role for anti-cancer immunity, (iv) indicate neuroendocrine and paracrine mediating mechanisms of stress- and surgery-induced promotion of cancer progression, focusing on the prominent role of catecholamines and prostaglandins through their impact on anti-cancer immunity, and through direct effects on the malignant tissue and its surrounding, (v) discuss the impact of different anesthetic approaches and other intra-operative procedures on immunity and cancer progression, and (vi) suggest prophylactic measures against the immunosuppressive and cancer promoting effects of surgery.
Collapse
Affiliation(s)
- Elad Neeman
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv 69978, Israel
| | | |
Collapse
|
20
|
Meron G, Tishler Y, Shaashua L, Rosenne E, Levi B, Melamed R, Gotlieb N, Matzner P, Sorski L, Ben-Eliyahu S. PGE2 suppresses NK activity in vivo directly and through adrenal hormones: effects that cannot be reflected by ex vivo assessment of NK cytotoxicity. Brain Behav Immun 2013; 28:128-38. [PMID: 23153554 PMCID: PMC3641317 DOI: 10.1016/j.bbi.2012.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 10/18/2012] [Accepted: 11/05/2012] [Indexed: 01/12/2023] Open
Abstract
Surgery can suppress in vivo levels of NK cell cytotoxicity (NKCC) through various mechanisms, including catecholamine-, glucocorticoid (CORT)-, and prostaglandin (PG)-mediated responses. However, PGs are synthesized locally following tissue damage, driving proinflammatory and CORT responses, while their systemic levels are often unaffected. Thus, we herein studied the role of adrenal factors in mediating in vivo effects of PGs on NKCC, using adrenalectomized and sham-operated F344 rats subjected to surgery or PGE(2) administration. In vivo and ex vivo approaches were employed, based on intravenous administration of the NK-sensitive MADB106 tumor line, and based on ex vivo assessment of YAC-1 and MADB106 target-line lysis. Additionally, in vitro studies assessed the kinetics of the impact of epinephrine, CORT, and PGE(2) on NKCC. The results indicated that suppression of NKCC by epinephrine and PGE(2) are short lasting, and cannot be evident when these compounds are removed from the in vitro assay milieu, or in the context of ex vivo assessment of NKCC. In contrast, the effects of CORT are long-lasting and are reflected in both conditions even after its removal. Marginating-pulmonary NKCC was less susceptible to suppression than circulating NKCC, when tested against the xenogeneic YAC-1 target line, but not against the syngeneic MADB106 line, which seems to involve different cytotoxicity mechanisms. Overall, these findings indicate that elevated systemic PG levels can directly suppress NKCC in vivo, but following laparotomy adrenal hormones mediate most of the effects of endogenously-released PGs. Additionally, the ex vivo approach seems limited in reflecting the short-lasting NK-suppressive effects of catecholamines and PGs.
Collapse
Affiliation(s)
- G Meron
- Neuroimmunology Research Unit, Department of Psychology, Tel-Aviv University, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Matzner P, Hazut O, Naim R, Shaashua L, Sorski L, Levi B, Sadeh A, Wald I, Bar-Haim Y, Ben-Eliyahu S. Resilience of the immune system in healthy young students to 30-hour sleep deprivation with psychological stress. Neuroimmunomodulation 2013; 20:194-204. [PMID: 23635771 DOI: 10.1159/000348698] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/30/2013] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Young adults often encounter sleep deprivation and stressful events. Both have been separately reported to modulate immunity, and occasionally they occur simultaneously. We assessed the combined effects of these conditions on immune competence in healthy students. METHODS Twenty-three participants (mean age 24 years; SD 1.86; 14 females) were exposed to 30 h of sleep deprivation during which they conducted physiological, social and cognitive tasks. The control group consisted of 18 participants (mean age 23.67 years; SD 1.46; 11 females). All participants underwent cognitive and psychological evaluations at 10:00 AM, followed by blood and saliva collection, 3 days before sleep deprivation induction and on the morning following it. Immune/endocrine measures included blood counts of lymphocytes, granulocytes, monocytes and natural killer (NK) cells; levels of several cell surface markers; NK cytotoxicity; plasma levels of interleukin (IL)-6, IL-10, dehydroepiandrosterone and neuropeptide Y, and plasma and salivary cortisol levels. RESULTS Although the experimental protocol significantly elevated state anxiety and psychological dissociation levels, no effects were evident in any of the immunological/endocrine indices. In contrast, expected sex differences in immune measures were found, including significantly higher NK cytotoxicity and monocyte counts in males, validating the integrity of the measurements. CONCLUSIONS The findings suggest resilience of the immune system to a combined sleep deprivation and stressful exposure in young adults, while previous studies reported immune perturbations following either of these conditions separately. These apparent contradictions might reflect differences in the study design or in the methodology used for immunological assessments, including the time of sample collection, the combination of sleep deprivation with stress and our in vivo assessment of cytokine levels.
Collapse
Affiliation(s)
- Pini Matzner
- School of Psychological Sciences, Tel Aviv University, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Neeman E, Zmora O, Ben-Eliyahu S. A new approach to reducing postsurgical cancer recurrence: perioperative targeting of catecholamines and prostaglandins. Clin Cancer Res 2012; 18:4895-902. [PMID: 22753587 DOI: 10.1158/1078-0432.ccr-12-1087] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Surgery is a crucial intervention in most cancer patients, but the perioperative period is characterized by increased risks for future outbreak of preexisting micrometastases and the initiation of new metastases-the major cause of cancer-related death. Here we argue that the short perioperative period is disproportionately critical in determining long-term recurrence rates, discuss the various underlying risk factors that act synergistically during this period, and assert that this time frame presents an unexplored opportunity to reduce long-term cancer recurrence. We then address physiologic mechanisms that underlie these risk factors, focusing on excess perioperative release of catecholamines and prostaglandins, which were recently shown to be prominent in facilitating cancer recurrence through their direct impact on the malignant tissue and its microenvironment, and through suppressing antimetastatic immunity. The involvement of the immune system is further discussed in light of accumulating evidence in cancer patients, and given the recent identification of endogenously activated unique leukocyte populations which, if not suppressed, can destroy autologous "immune-resistant" tumor cells. We then review animal studies and human correlative findings, suggesting the efficacy of blocking catecholamines and/or prostaglandins perioperatively, limiting metastasis and increasing survival rates. Finally, we propose a specific perioperative pharmacologic intervention in cancer patients, based on simultaneous β-adrenergic blockade and COX-2 inhibition, and discuss specific considerations for its application in clinical trials, including our approved protocol. In sum, we herein present the rationale for a new approach to reduce long-term cancer recurrence by using a relatively safe, brief, and inexpensive intervention during the perioperative period.
Collapse
Affiliation(s)
- Elad Neeman
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv, Israel
| | | | | |
Collapse
|
23
|
Neeman E, Shaashua L, Benish M, Page GG, Zmora O, Ben-Eliyahu S. Stress and skin leukocyte trafficking as a dual-stage process. Brain Behav Immun 2012; 26:267-76. [PMID: 21963875 PMCID: PMC3253878 DOI: 10.1016/j.bbi.2011.09.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2011] [Revised: 09/04/2011] [Accepted: 09/15/2011] [Indexed: 10/17/2022] Open
Abstract
Stress responses are known to modulate leukocyte trafficking. In the skin, stress was reported both to enhance and reduce skin immunity, and the chronicity of stress exposure was suggested as a key determining factor. We here propose a dual-stage hypothesis, suggesting that stress, of any duration, reduces skin immunity during its course, while its cessation is potentially followed by a period of enhanced skin immunity. To start testing this hypothesis, rats were subcutaneously implanted with sterile surgical sponges for four-hours, during or after exposure to one of several acute stress paradigms, or to a chronic stress paradigm. Our findings, in both males and females, indicate that numbers of sponge-infiltrating leukocytes, and their specific subsets, were reduced during acute or chronic stress, and increased after stress cessation. Studying potential mediating mechanisms of the reduction in leukocyte numbers during acute stress, we found that neither adrenalectomy nor the administration of beta-adrenergic or glucocorticoid antagonists prevented this reduction. Additionally, administration of corticosterone or epinephrine to adrenalectomized rats did not impact skin leukocyte numbers, whereas, in the blood, these treatments did affect numbers of leukocytes and their specific subsets, as was also reported previously. Overall, our findings support the proposed dual-stage hypothesis, which can be evolutionally rationalized and accounts for most of the apparent inconsistencies in the literature regarding stress and skin immunity. Other aspects of the hypothesis should be tested, also using additional methodologies, and its predictions may bear clinical significance in treatment of skin disorders related to hyper- or hypo-immune function.
Collapse
Affiliation(s)
- Elad Neeman
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv, Israel
| | - Lee Shaashua
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv, Israel
| | - Marganit Benish
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv, Israel
| | - Gayle G. Page
- Johns Hopkins University School of Nursing, 525 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Oded Zmora
- Department of Surgery and Transplantation, Sheba Medical Center, 52621 Tel Hashomer, Israel
| | - Shamgar Ben-Eliyahu
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
24
|
Chen Z, Cao J, Liao X, Ke J, Zhu S, Zhao P, Qi Z. Plasmids Enriched with CpG Motifs Activate Human Peripheral Blood Mononuclear Cells In Vitro and Enhance Th-1 Immune Responses to Hepatitis B Surface Antigen in Mice. Viral Immunol 2011; 24:199-209. [DOI: 10.1089/vim.2010.0116] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Zhihui Chen
- Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jie Cao
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai, China
| | - Xiaoling Liao
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai, China
| | - Jinshan Ke
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai, China
| | - Shiying Zhu
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai, China
| | - Ping Zhao
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai, China
| | - Zhongtian Qi
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai, China
| |
Collapse
|
25
|
Levi B, Benish M, Goldfarb Y, Sorski L, Melamed R, Rosenne E, Ben-Eliyahu S. Continuous stress disrupts immunostimulatory effects of IL-12. Brain Behav Immun 2011; 25:727-35. [PMID: 21277367 PMCID: PMC3081380 DOI: 10.1016/j.bbi.2011.01.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 01/05/2011] [Accepted: 01/23/2011] [Indexed: 11/28/2022] Open
Abstract
Immune stimulation by biological response modifiers is a common approach in tumor immunotherapy. IL-12 was found effective in various animal studies, but clinical trials showed limited success. However, among other differences, animal models do not simulate psychological or physiological stress while employing IL-12, whereas cancer patients often experience distress while treated with immunostimulants. Thus, in the current study we assessed the impact of continuous stress on the efficacy of IL-12 immunostimulation. F344 rats were subjected to a pharmacological stress paradigm (continuous administration of a β-adrenergic agonist) or to a 20 h behavioral stress paradigm (wet cage exposure) commencing 2h before IL-12 administration. Twenty-six hours after stress initiation, we studied indices known to reflect IL-12 immunostimulatory impacts, including NK cell numbers and activity in different immune compartments, and in vivo resistance to MADB106 lung tumor colonization. The results indicated that both the pharmacological and behavioral stress paradigms significantly reduced the increase in the number and activity of marginating-pulmonary NK cells evident in non-stressed IL-12 treated animals. Additionally, stressed animals exhibited a lower IL-12-induced improvement of MADB106 lung clearance, an in vivo index that markedly depends on total marginating-pulmonary NK activity. These deleterious effects of stress were more prominent in males than in females. Overall, the findings demonstrate that prolonged stress exposure can disrupt the efficacy of simultaneous immunostimulatory treatments, irrespective of stress effects on baseline immune measures. Neuroendocrine and cellular mediating mechanisms are yet unknown, but the potential clinical ramifications of these findings warrant consideration in clinical trials employing immunostimulatory agents.
Collapse
MESH Headings
- Adenocarcinoma/complications
- Adenocarcinoma/immunology
- Adjuvants, Immunologic/administration & dosage
- Adrenergic beta-2 Receptor Agonists
- Analysis of Variance
- Animals
- Chronic Disease
- Corticosterone/blood
- Epinephrine/blood
- Female
- Interleukin-12/administration & dosage
- Interleukin-12/immunology
- Killer Cells, Natural/cytology
- Killer Cells, Natural/immunology
- Lung Neoplasms/complications
- Lung Neoplasms/immunology
- Male
- Metaproterenol
- Neoplasms, Experimental/complications
- Neoplasms, Experimental/immunology
- Rats
- Rats, Inbred F344
- Sex Factors
- Stress, Psychological/blood
- Stress, Psychological/chemically induced
- Stress, Psychological/complications
- Stress, Psychological/immunology
Collapse
Affiliation(s)
- Ben Levi
- Neuroimmunology Research Unit, Department of Psychology, Tel-Aviv University, Tel-Aviv 69978, Israel
| | | | | | | | | | | | | |
Collapse
|
26
|
Goldfarb Y, Levi B, Sorski L, Frenkel D, Ben-Eliyahu S. CpG-C immunotherapeutic efficacy is jeopardized by ongoing exposure to stress: potential implications for clinical use. Brain Behav Immun 2011; 25:67-76. [PMID: 20656015 PMCID: PMC2991469 DOI: 10.1016/j.bbi.2010.07.242] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 07/16/2010] [Accepted: 07/16/2010] [Indexed: 12/17/2022] Open
Abstract
Bi-directional influences between stress hormones and immune responses have been repeatedly documented, however, in the clinical setting they are rarely considered when immunotherapeutic approaches are used or studied in patients. As some immunotherapeutic treatments have shown great potential in animal models but have had limited success in patients, we hypothesize that ongoing psychological and physiological stress responses in patients, which do not characterize the setting of animal studies, contribute to this discrepancy. In the current study we examined the interaction between ongoing water stress and CpG-C immunotherapy to determine whether stress that precedes immunotherapy can modulate the efficacy of CpG-C immunostimulation. C57BL/6 mice were exposed to water stress or served as controls. Two hours following the commencement of the stress protocol animals were injected with CpG-C, non-CpG, or PBS, and sacrificed 1, 4 or 12h thereafter. We found that in CpG-C-treated animals stress eliminated the elevation of plasma IL-12, and synergistically elevated corticosterone levels. Furthermore, stress markedly reduced the total number of myeloid (33D1(+)), plasmacytoid (mPDCA-1(+)) and plasmacytoid-derived (33D1(+)mPDCA-1(+)) dendritic cells in CpG-C-treated animals, as well as the numbers of these cell sub-types expressing CD11b, CD80 and CD69. These changes were more dramatic in the blood than in the spleen. Overall, these findings indicate that under no-stress conditions CpG-C induces a robust immune response, which is significantly diminished when immunostimulation is attempted during ongoing stress. If these findings hold in humans, potential prophylactic treatments should be found to limit the deleterious effects of ongoing stress on the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Yael Goldfarb
- Neuroimmunology Research Unit, Department of Psychology, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Ben Levi
- Neuroimmunology Research Unit, Department of Psychology, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Liat Sorski
- Neuroimmunology Research Unit, Department of Psychology, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Dan Frenkel
- Department of Neurobiology, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Shamgar Ben-Eliyahu
- Neuroimmunology Research Unit, Department of Psychology, Tel-Aviv University, Tel-Aviv 69978, Israel
| |
Collapse
|
27
|
Benish M, Ben-Eliyahu S. Surgery as a double-edged sword: a clinically feasible approach to overcome the metastasis-promoting effects of surgery by blunting stress and prostaglandin responses. Cancers (Basel) 2010; 2:1929-51. [PMID: 24281210 PMCID: PMC3840453 DOI: 10.3390/cancers2041929] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 11/14/2010] [Accepted: 11/23/2010] [Indexed: 12/22/2022] Open
Abstract
Surgery remains an essential therapeutic approach for most solid malignancies, including breast cancer. However, surgery also constitutes a risk factor for promotion of pre-existing micrometastases and the initiation of new metastases through several mechanisms, including the release of prostaglandins and stress hormones (e.g., catecholamines and glucocorticoids). However, the perioperative period also presents an opportunity for cell mediated immunity (CMI) and other mechanisms to eradicate or control minimal residual disease, provided that the deleterious effects of surgery are minimized. Here, we discuss the key role of endogenous stress hormones and prostaglandins in promoting the metastatic process through their direct impact on malignant cells, and through their deleterious impact on anti-cancer CMI. We further discuss the effects of anesthetic techniques, the extent of surgery, pain alleviation, and timing within the menstrual cycle with respect to their impact on tumor recurrence and physiological stress responses. Last, we suggest an attractive perioperative drug regimen, based on a combination of a cyclooxygenase (COX)-2 inhibitor and a β-adrenergic blocker, which we found effective in attenuating immune suppression and the metastasis-promoting effects of surgery in several tumor models. This regimen is clinically applicable, and could potentially promote disease free survival in patients operated for breast and other types of cancer.
Collapse
Affiliation(s)
- Marganit Benish
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv 69978, Israel.
| | | |
Collapse
|