Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
2.
BMC Biol ; 18(1): 163, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33158447

ABSTRACT

BACKGROUND: Numerous case studies have reported spontaneous regression of recognized metastases following primary tumor excision, but underlying mechanisms are elusive. Here, we present a model of regression and latency of metastases following primary tumor excision and identify potential underlying mechanisms. RESULTS: Using MDA-MB-231HM human breast cancer cells that express highly sensitive luciferase, we monitored early development stages of spontaneous metastases in BALB/c nu/nu mice. Removal of the primary tumor caused marked regression of micro-metastases, but not of larger metastases, and in vivo supplementation of tumor secretome diminished this regression, suggesting that primary tumor-secreted factors promote early metastatic growth. Correspondingly, MDA-MB-231HM-conditioned medium increased in vitro tumor proliferation and adhesion and reduced apoptosis. To identify specific mediating factors, cytokine array and proteomic analysis of MDA-MB-231HM secretome were conducted. The results identified significant enrichment of angiogenesis, growth factor binding and activity, focal adhesion, and metalloprotease and apoptosis regulation processes. Neutralization of MDA-MB-231HM-secreted key mediators of these processes, IL-8, PDGF-AA, Serpin E1 (PAI-1), and MIF, each antagonized secretome-induced proliferation. Moreover, their in vivo simultaneous blockade in the presence of the primary tumor arrested the development of micro-metastases. Interestingly, in the METABRIC cohort of breast cancer patients, elevated expression of Serpin E1, IL-8, or the four factors combined predicted poor survival. CONCLUSIONS: These results demonstrate regression and latency of micro-metastases following primary tumor excision and a crucial role for primary tumor secretome in promoting early metastatic growth in MDA-MB-231HM xenografts. If generalized, such findings can suggest novel approaches to control micro-metastases and minimal residual disease.


Subject(s)
Breast Neoplasms/surgery , Cell Proliferation , Neoplasm Regression, Spontaneous/physiopathology , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Proteomics
3.
Cancer Immunol Immunother ; 69(10): 2021-2031, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32405793

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.


Subject(s)
Colonic Neoplasms/prevention & control , Killer Cells, Natural/drug effects , Liver Neoplasms/prevention & control , Oligodeoxyribonucleotides/administration & dosage , Animals , Apoptosis , Cell Proliferation , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Perioperative Period , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Brain Behav Immun ; 80: 170-178, 2019 08.
Article in English | MEDLINE | ID: mdl-30851377

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.


Subject(s)
Neoplasm Metastasis/immunology , Perioperative Period/methods , Stress, Psychological/immunology , Animals , Cell Line, Tumor , Female , Laparotomy/adverse effects , Laparotomy/psychology , Male , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/physiopathology , Neoplasms/metabolism , Neoplasms/surgery , Rats , Rats, Inbred F344 , Surgical Procedures, Operative/adverse effects , Surgical Procedures, Operative/psychology , Treatment Outcome
5.
Endocrinology ; 159(1): 248-259, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29059290

ABSTRACT

It is assumed that after complete bilateral adrenalectomy (ADX), no adrenal tissue will redevelop and adrenal hormone levels will remain low and unaffected by stress. However, anecdotal observations in animals and in patients suggest that under some unknown circumstances the opposite can occur. Herein, we studied whether adrenalectomized rats can develop an alternative source of systemic corticosterone after complete bilateral ADX with minimal replacement therapy. Male and female rats underwent either a standard ADX, in which the glands were removed with minimal surrounding adipose tissue, or an extensive ADX, in which glands were removed with most surrounding adipose tissue. Excised glands were histologically tested for completeness, and corticosterone replacement was nullified within 1 to 3 weeks postoperatively. In four experiments and in both excision approaches, some rats gradually reestablished baseline corticosterone levels and stress response in a time-dependent manner, but differences were observed in the reestablishing rates: 80% in standard ADX vs 20% in extensive ADX. Upon searching for the source of corticosterone secretion, we were surprised to find functional macroscopic foci of adrenocortical tissue without medullary tissue, mostly proximal to the original location. Chronic stress accelerated corticosterone level reestablishment. We hypothesized that underlying this phenomenon were preexisting ectopic microscopic foci of adrenocortical-like tissue or a few adrenal cells that were pre-embedded in surrounding tissue or detached from the excised gland upon removal. We concluded that adrenalectomized animals may develop compensatory mechanisms and suggest that studies employing ADX consider additional corticosterone supplementation, minimize stress, and verify the absence of circulating corticosterone.


Subject(s)
Adrenal Glands/physiology , Adrenalectomy/adverse effects , Aging , Corticosterone/administration & dosage , Hormone Replacement Therapy , Regeneration , Stress, Physiological , Adrenal Cortex/drug effects , Adrenal Cortex/metabolism , Adrenal Cortex/physiology , Adrenal Cortex/surgery , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Adrenal Glands/surgery , Adrenalectomy/methods , Animals , Corticosterone/blood , Corticosterone/metabolism , Female , Injections, Subcutaneous , Male , Margins of Excision , Postoperative Period , Random Allocation , Rats, Inbred F344 , Rats, Inbred Lew , Regeneration/drug effects , Restraint, Physical/adverse effects , Survival Analysis
6.
Brain Behav Immun ; 58: 91-98, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27235931

ABSTRACT

Liver metastases are a major cause of colorectal cancer death, and the perioperative period is believed to critically affect the metastatic process. Here we tested whether blocking excess release of catecholamines and prostaglandins during surgical procedures of different extent can reduce experimental liver metastasis of the syngeneic CT26 colon cancer in female and male BALB/c mice. Animals were either treated with the beta-blocker, propranolol, the COX-2 inhibitor, etodolac, both drugs, or vehicle. The role of NK cells in controlling CT26 hepatic metastasis and in mediating the effect of the drugs was assessed by in vivo depletion or stimulation of NK cells, using anti-asialo GM1 or CpG-C, respectively. Surgical extent was manipulated by adding laparotomy to small incision, extending surgical duration, and enabling hypothermia. The results indicated that combined administration of propranolol and etodolac, but neither drug alone, significantly improved host resistance to metastasis. These beneficial effects occurred in both minor and extensive surgeries, in both sexes, and in two tumor inoculation approaches. NK cell-mediated anti-CT26 activity is involved in mediating the beneficial effects of the drugs. Specifically, CpG-C treatment, known to profoundly activate mice marginating-hepatic NK cytotoxicity, reduced CT26 hepatic metastases; and NK-depletion increased metastases and prevented the beneficial effects of the drugs. Overall, given prevalent perioperative psychological and physiological stress responses in patients, and ample prostaglandin release by colorectal tumors and injured tissue, propranolol and etodolac could be tested clinically in laparoscopic and open colorectal surgeries, attempting to reduce patients' metastatic disease.


Subject(s)
Adrenergic beta-Agonists/administration & dosage , Antineoplastic Agents/administration & dosage , Colonic Neoplasms/surgery , Cyclooxygenase 2 Inhibitors/administration & dosage , Etodolac/administration & dosage , Liver Neoplasms/prevention & control , Propranolol/administration & dosage , Animals , Cell Line, Tumor , Female , Killer Cells, Natural/metabolism , Laparotomy , Liver Neoplasms/secondary , Male , Mice, Inbred BALB C , Minor Surgical Procedures
7.
Int J Cancer ; 138(7): 1754-64, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26453448

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.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antineoplastic Agents/pharmacology , Glucosides/pharmacology , Lipid A/pharmacology , Neoplasm Metastasis/drug therapy , Neoplasms, Experimental/pathology , Toll-Like Receptor 4/agonists , Animals , Cell Line, Tumor , Female , Flow Cytometry , Male , Mice , Mice, Inbred BALB C , Perioperative Period , Rats , Rats, Inbred F344
8.
Brain Behav Immun ; 45: 277-86, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25546569

ABSTRACT

In vitro and ex vivo studies assessing the impact of stress hormones on immune competence commonly replace the natural milieu of leukocytes with an artificial medium, excluding plasma factors, hormones, and cytokines. Given prevalent inconsistencies between in vitro, ex vivo, and in vivo findings, we studied whether such procedures could yield misleading outcomes regarding the impact of stress hormones on NK cell cytotoxicity (NKCC), using fresh human whole blood samples. We found that in the presence of plasma 10-30-fold higher concentrations of cortisol, epinephrine, and prostaglandin-E2 (PGE2) were required to reach suppression levels evident in the context of artificial medium. Importantly, whereas the NK suppressive effects of PGE2 occurred immediately and remained stable upon prolonged exposure, the suppressive effects of cortisol slowly increased over time. Last, to simulate the exclusion of stress factors in the ex vivo approach, we subjected whole blood to stress hormones (as occurs in vivo), and abruptly removed them. We found that the effects of epinephrine and PGE2 quickly disappeared, while the effects of cortisol persisted. Overall, these findings demonstrate the potential misleading nature of in vitro and ex vivo procedures, and specifically suggest that (i) the common in vitro findings of profound suppression of NKCC by stress hormones are overestimation of their direct effects expected in vivo; and (ii) the common ex vivo approach cannot reflect the direct in vivo suppressive effects of epinephrine and PGE2 on NKCC, while inflating the effects of glucocorticoids. Some of these fallacies may be circumvented by using non-delayed whole blood NKCC assays in humans.


Subject(s)
Cytotoxicity, Immunologic/immunology , Dinoprostone/immunology , Epinephrine/immunology , Hydrocortisone/immunology , Killer Cells, Natural/immunology , Plasma/immunology , Stress, Psychological/immunology , Humans , In Vitro Techniques
9.
Psychoneuroendocrinology ; 42: 11-23, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24636497

ABSTRACT

IL-12 is a prominent Th1 differentiator and leukocyte activator. Ample studies showed suppression of IL-12 production by numerous stress factors, including prostaglandins, catecholamines, glucocorticoids, and opioids, but did so in vitro and in the context of artificial leukocyte activation, not simulating the in vivo setting. In a recent study we reported in vivo suppression of plasma IL-12 levels by behavioral stress and surgery. The current study aims to elucidate neuroendocrine mechanisms underlying this phenomenon in naïve F344 rats. To this end, both adrenalectomy and administration of specific antagonists were used, targeting the aforementioned stress factors. The results indicated that corticosterone and prostaglandins are prominent mediators of the IL-12-suppressing effects of stress and surgery, apparently through directly suppressing leukocyte IL-12 production. Following surgery, endogenous prostaglandins exerted their effects mainly through elevating corticosterone levels. Importantly, stress-induced release of epinephrine or opioids had no impact on plasma IL-12 levels, while pharmacological administration of epinephrine reduced plasma IL-12 levels by elevating corticosterone levels. Last, a whole blood in vitro study indicated that prostaglandins and corticosterone, but not epinephrine, suppressed IL-12 production in non-stimulated leukocytes, and only corticosterone did so in the context of CpG-C-induced IL-12 production. Overall, the findings reiterate the notion that results from in vitro or pharmacological in vivo studies cannot indicate the effects of endogenously released stress hormones under stress/surgery conditions. Herein, corticosterone and prostaglandins, but not catecholamines or opioids, were key mediators of the suppressive effect of stress and surgery on in vivo plasma IL-12 levels in otherwise naïve animals.


Subject(s)
Corticosterone/blood , Interleukin-12/blood , Prostaglandins/blood , Stress, Physiological/physiology , Stress, Psychological/blood , Adrenalectomy , Animals , Epinephrine/pharmacology , Male , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Rats , Rats, Inbred F344
10.
Immunol Res ; 58(1): 28-39, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24132552

ABSTRACT

To test whether marginating-pulmonary (MP) leukocytes in mice have a unique potential to identify and destroy aberrant circulating cells, we compared MP to circulating leukocytes with respect to natural killer (NK) cytotoxicity, proinflammatory characteristics, molecular determinants of activation, and response to IL-12 immunostimulation. Cytotoxicity was assessed employing the YAC-1, B16F10, and 3LL target lines. C57BL/6 mice were injected with either saline or murine IL-12 (0.1 or 0.5 µg/mouse), either once or three times 48-h apart. Twenty-four hours after last injection, cardiac blood was withdrawn and MP leukocytes were collected by forced lung perfusion. NK cytotoxicity, cellular composition, and surface molecular markers were studied. MP leukocytes exhibited greater NK cytotoxicity than circulating leukocytes against the syngeneic B16F10 and 3LL tumor lines, but not against the allogeneic YAC-1 line. NKG2D and IL-12 receptor expression predicted NK cytotoxicity in circulating leukocytes, but not in MP leukocytes. IFNγ-receptor, IL-12-receptor, CD69, CD11a, and CD11b showed different patterns of expression in the two leukocyte populations, suggesting pro-inflammatory characteristics of the MP compartment. IL-12 stimulation caused differential effects on these markers and also elevated cytotoxicity in both compartments, but in different effector: target ratio-dependent patterns. MP leukocytes may play a critical role in eliminating aberrant circulating cells due to their enhanced NK cytotoxicity and given their strategic location in the lungs vasculature, which forces physical interactions with all circulating aberrant cells. MP-NK cells are unique in their cytotoxic mechanisms against syngeneic targets and in their activation profile and response to immunostimulatory agents.


Subject(s)
Interleukin-12/immunology , Killer Cells, Natural/immunology , Leukocytes/immunology , Lung/immunology , Animals , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , CD11a Antigen/immunology , CD11b Antigen/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Flow Cytometry , Killer Cells, Natural/cytology , Lectins, C-Type/immunology , Leukocytes/cytology , Lung/cytology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Receptors, Interferon/immunology , Receptors, Interleukin-12/immunology
11.
Brain Behav Immun ; 37: 207-19, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24333572

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.


Subject(s)
Catecholamines/physiology , Cytotoxicity, Immunologic , Glucocorticoids/physiology , Killer Cells, Natural/immunology , Prostaglandins/physiology , Stress, Physiological , Adrenalectomy , Animals , Catecholamines/metabolism , Cell Line, Tumor , Corticosterone/pharmacokinetics , Corticosterone/pharmacology , Female , Glucocorticoids/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Male , Prostaglandins/metabolism , Rats , Rats, Inbred F344
12.
PLoS One ; 6(4): e19246, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21559428

ABSTRACT

In leukemia patients, stress and anxiety were suggested to predict poorer prognosis. Oncological patients experience ample physiological and psychological stress, potentially leading to increased secretion of stress factors, including epinephrine, corticosteroids, and prostaglandins. Here we tested whether environmental stress and these stress factors impact survival of leukemia-challenged rats, and studied mediating mechanisms. F344 rats were administered with a miniscule dose of 60 CRNK-16 leukemia cells, and were subjected to intermittent forced swim stress or to administration of physiologically relevant doses of epinephrine, prostaglandin-E(2) or corticosterone. Stress and each stress factor, and/or their combinations, doubled mortality rates when acutely applied simultaneously with, or two or six days after tumor challenge. Acute administration of the ß-adrenergic blocker nadolol diminished the effects of environmental stress, without affecting baseline survival rates. Prolonged ß-adrenergic blockade or COX inhibition (using etodolac) also increased baseline survival rates, possibly by blocking tumor-related or normal levels of catecholamines and prostaglandins. Searching for mediating mechanisms, we found that each of the stress factors transiently suppressed NK activity against CRNK-16 and YAC-1 lines on a per NK basis. In contrast, the direct effects of stress factors on CRNK-16 proliferation, vitality, and VEGF secretion could not explain or even contradicted the in vivo survival findings. Overall, it seems that environmental stress, epinephrine, and prostaglandins promote leukemia progression in rats, potentially through suppressing cell mediated immunity. Thus, patients with hematological malignancies, which often exhibit diminished NK activity, may benefit from extended ß-blockade and COX inhibition.


Subject(s)
Dinoprostone/metabolism , Epinephrine/metabolism , Killer Cells, Natural/cytology , Leukemia/pathology , Animals , Cell Line, Tumor , Corticosterone/metabolism , Disease Models, Animal , Disease Progression , Female , Male , Rats , Rats, Inbred F344 , Stress, Psychological , Swimming , Time Factors , Treatment Outcome
13.
Brain Behav Immun ; 25(4): 727-35, 2011 May.
Article in English | MEDLINE | ID: mdl-21277367

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 ß-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.


Subject(s)
Adenocarcinoma/immunology , Adjuvants, Immunologic/administration & dosage , Interleukin-12/immunology , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Stress, Psychological/immunology , Adenocarcinoma/complications , Adrenergic beta-2 Receptor Agonists , Analysis of Variance , Animals , Chronic Disease , Corticosterone/blood , Epinephrine/blood , Female , Interleukin-12/administration & dosage , Killer Cells, Natural/cytology , Lung Neoplasms/complications , Male , Metaproterenol , Neoplasms, Experimental/complications , Neoplasms, Experimental/immunology , Rats , Rats, Inbred F344 , Sex Factors , Stress, Psychological/blood , Stress, Psychological/chemically induced , Stress, Psychological/complications
14.
Brain Behav Immun ; 24(6): 952-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20362661

ABSTRACT

BACKGROUND: A unique opportunity to eradicate cancer is presented immediately after the excision of the primary tumor, but surgical procedures often induce the release of immunosuppressing factors that render cell mediated immunity ineffective. Here we tested the hypothesis that integration of peri-operative immunostimulation and blockade of immunosuppression could synergistically improve post-operative anti-metastatic immunity and long-term survival. METHODS: Two syngeneic tumor models in F344 rats were employed, studying post-operative tumor progression. In the first model, survival following laparotomy and CRNK-16 leukemia was studied. Rats were peri-operatively treated with the immuno-stimulant poly I-C (5x0.2 mg/kg/inj), with catecholamine- and prostaglandin-blockers (shown to prevent post-operative immunosuppression: 4.5 mg/kg nadolol, 4 mg/kg indomethacin), with both interventions, or with neither. Long-term survival was assessed thereafter. The second model used the MADB106 mammary adenocarcinoma, assessing its lung tumor retention (LTR) following i.v. inoculation, as well as host marginating-pulmonary NK numbers and activity against this tumor. IL-12 was employed for immunostimulation (4x1.5 microg/kg/inj), with and without the above blockers. RESULTS: Post-operative CRNK-16 survival rates were significantly improved only by the integrated approach of immune stimulation and endocrine blockers. Post-operative MADB106 LTR was additively reduced by the two interventions. Importantly, while IL-12 increased pulmonary NK cytotoxicity against MADB106, surgery markedly suppressed this cytotoxicity in both IL-12 and vehicle treated animals. The blockers prevented this suppression per lung and per single NK cell. CONCLUSIONS: Immunostimulation could be rendered ineffective post-operatively due to immunosuppression; therefore integrating endocrine-blocker therapies into the realm of peri-operative immunotherapy could optimize immune control over residual disease, potentially improving clinical outcomes.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immune Tolerance/physiology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/surgery , Postoperative Complications/immunology , Postoperative Complications/pathology , Adrenergic beta-Antagonists/pharmacology , Animals , Catecholamines/antagonists & inhibitors , Cell Line, Tumor , Cyclooxygenase Inhibitors/pharmacology , Disease Progression , Flow Cytometry , Hormone Antagonists/pharmacology , Indomethacin/pharmacology , Interleukin-12/pharmacology , Killer Cells, Natural , Laparotomy , Lung Neoplasms/pathology , Nadolol/pharmacology , Poly I-C/pharmacology , Prostaglandin Antagonists/pharmacology , Rats , Rats, Inbred F344 , Survival
15.
J Immunol ; 184(5): 2449-57, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20124103

ABSTRACT

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


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Lewis Lung/surgery , Melanoma, Experimental/surgery , Postoperative Complications/prevention & control , Adrenergic beta-Antagonists/administration & dosage , Amputation, Surgical/adverse effects , Animals , CD11a Antigen/metabolism , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Cyclooxygenase 2 Inhibitors/administration & dosage , Etodolac/administration & dosage , Fas Ligand Protein/metabolism , Female , Interleukin-12/administration & dosage , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Laparotomy/adverse effects , Male , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Poly I-C/administration & dosage , Postoperative Complications/etiology , Postoperative Complications/mortality , Propranolol/administration & dosage , Survival Rate
16.
J Immunother ; 33(1): 16-29, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19952959

ABSTRACT

A significant role has been indicated for cellular immunity in controlling circulating cancer cells, but most autologous tumor cells seem resistant, in vitro, to natural killer cell (NKC) and cytotoxic T lymphocytes cytotoxicity. Addressing this apparent contradiction, we recently identified a unique leukocyte population, marginating-pulmonary (MP)-leukocytes, which exhibit potent natural killer (NK) cytotoxicity. Here, we characterize the MP-compartment in naive and immunostimulated rats, and assessed its cytotoxicity against "NK-resistant" tumors cells. Animals were treated with poly I-C (3x0.2 mg/kg) or saline, and circulating-leukocytes and MP-leukocytes were collected and analyzed in terms of cellular composition, cellular activation markers, and NK cytotoxicity of leukocytes and purified NKCs. Compared with circulating-leukocytes, MP-leukocytes showed greater proportion of granulocytes, monocytes, NKCs, and large NKCs; higher expression of activation and adhesion markers (CD25, CD11a, CD11b, and NKR-P1, IFN-gamma); and elevated NK cytotoxicity of leukocytes and purified NKCs against several syngeneic and xenogeneic NK-resistant target cells (from both F344 and BDX inbred rats). In immunostimulated animals (treated with poly I-C), but not in naive animals, purified NKCs from the MP-compartment showed markedly superior cytotoxicity, suggesting that poly I-C immunostimulation uniquely affect MP-NKCs, and that in naive animals other MP-leukocytes support NK cytotoxicity. Overall, the results suggest that the MP-compartment is characterized by a continuous activated inflammatory microenvironment uniquely affected by immunostimulation. If similarly potent MP-NKCs exist in patients, then circulating autologous tumor cells that are considered "NK-resistant" could actually be controlled by MP-NKCs. Innate immunity may assume greater role in controlling malignant spread, especially after immunostimulation.


Subject(s)
Cytotoxicity, Immunologic/immunology , Inflammation/immunology , Killer Cells, Natural/immunology , Lung/immunology , Neoplasms, Experimental/immunology , Animals , Cell Separation , Flow Cytometry , Interferon Inducers/immunology , Interferon Inducers/pharmacology , Killer Cells, Natural/cytology , Lung/cytology , Male , Phenotype , Poly I-C/immunology , Poly I-C/pharmacology , Rats , Rats, Inbred F344
17.
J Immunother ; 32(3): 280-91, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19242372

ABSTRACT

Suppression of natural killer (NK) cell activity is common after stress, has been reported to predict malignant recurrence in cancer patients, and was shown to underlie metastatic dissemination in animal models. We have previously reported that catecholamines play a major role in NK cell suppression, particularly in the context of physiologic stress and surgery. In the current study using Fisher 344 rats, we examined the prophylactic use of different regimens of type-C CpG oligodeoxynucleotides (CpG-C ODN) on NK activity and metastatic dissemination in the context of pharmacologic stress (using metaproterenol for beta-adrenoceptor stimulation). Our results indicated that the beneficial effects of CpG-C ODN were more profound under pharmacologic stress than under baseline conditions. A bolus of CpG-C ODN (330 microg/kg, intraperitoneally) 24 hours before metaproterenol-challenge was most effective at reducing lung tumor retention of an experimental syngeneic mammary adenocarcinoma (MADB106), although having no observable side effects. Depletion of NK cells revealed their key role in improving baseline levels of resistance to metastatic dissemination after CpG-C ODN administration. When NK cell cytotoxicity was assessed in the circulation and the marginating-pulmonary immune compartments, we found that CpG-C ODN protected individual NK cells from metaproterenol-induced suppression in both compartments. Moreover, in the critical marginating-pulmonary compartment, CpG-C ODN also elevated baseline cytotoxicity per NK cell against MADB106 tumor cells, and increased NK cell numbers in nonstressed rats. Overall, prophylactic CpG-C ODN treatment can improve immunocompetence and potentially reduce metastatic dissemination, especially in clinical settings characterized by enhanced sympathetic stress responses.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cytotoxicity, Immunologic/drug effects , Killer Cells, Natural/drug effects , Neoplasm Metastasis/immunology , Oligodeoxyribonucleotides/pharmacology , Receptors, Adrenergic, beta/immunology , Animals , Cell Line, Tumor , Immunosuppression Therapy , Killer Cells, Natural/immunology , Mammary Neoplasms, Animal/immunology , Metaproterenol/pharmacology , Rats , Rats, Inbred F344 , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/metabolism
18.
Brain Behav Immun ; 23(5): 611-21, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18951972

ABSTRACT

Inflammation is implicated in several medical conditions that are sexually dimorphic, including depression, cardiovascular diseases, autoimmunity, and presumably cancer progression. Here we studied the effects of the proinflammatory agent, LPS, on MADB106 lung tumor retention (LTR), and sought to elucidate underlying mechanisms and sexual dimorphism. F344 male and female rats were administered with LPS (0.001-1mg/kg i.v.) simultaneously with tumor cell inoculation, and treated with a beta-blocker (nadolol, 0.2-0.3mg/kg s.c.), a COX inhibitor (indomethacin, 4mg/kg s.c.) or both drugs. To study the role of NK cells, numbers and cytotoxicity of marginating-pulmonary NK cells were studied, and selective in vivo NK-depletion was employed. Serum levels of corticosterone, IL-6, and TNF-alpha were also assessed. The findings indicated that LPS increased LTR in both sexes, but 10-fold higher doses were needed in females to reach the increase evident in males. Additionally, nadolol and indomethacin reduced the effects of LPS, more so in males. In vivo NK-depletion and ex vivo NK activity studies suggested that LPS affected LTR through both NK-independent and NK-dependent mechanisms, the latter mediated through prostaglandin release in males. Corticosterone, IL-6, and TNF-alpha responses to LPS were sexually dimorphic, but were not associated with LPS or drugs' impacts on LTR. Overall, our findings demonstrate sexual dimorphism in LPS-induced elevated susceptibility to MADB106 experimental metastasis, and in potential humoral underlying mechanisms. Further studies are needed to elucidate additional immunological and non-immunological mediators of these dimorphisms, as well as to assess their involvement in other sexually dimorphic pathologies that are associated with inflammation.


Subject(s)
Adenocarcinoma/secondary , Catecholamines/physiology , Lipopolysaccharides/toxicity , Lung Neoplasms/secondary , Prostaglandins/physiology , Sex Characteristics , Adenocarcinoma/blood , Adenocarcinoma/prevention & control , Adrenergic beta-Antagonists/pharmacology , Animals , Cell Line, Tumor/transplantation , Corticosterone/blood , Cyclooxygenase Inhibitors/pharmacology , Cytotoxicity, Immunologic , Female , Indomethacin/pharmacology , Interleukin-6/blood , Killer Cells, Natural/immunology , Lung Neoplasms/blood , Lung Neoplasms/prevention & control , Male , Mammary Neoplasms, Experimental/pathology , Nadolol/pharmacology , Rats , Rats, Inbred F344 , Tumor Necrosis Factor-alpha/analysis
19.
Anesthesiology ; 109(6): 989-97, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19034095

ABSTRACT

BACKGROUND: In cancer patients, allogeneic blood transfusion is associated with poorer prognosis, but the independent effect of the transfusion is controversial. Moreover, mediating mechanisms underlying the alleged cancer-promoting effects of blood transfusion are unknown, including the involvement of donors' leukocytes, erythrocytes, and soluble factors. METHOD: Two syngeneic tumor models were used in Fischer 344 rats, the MADB106 mammary adenocarcinoma and the CRNK-16 leukemia. Outcomes included host ability to clear circulating cancer cells, and host survival rates. The independent impact of blood transfusion was assessed, and potential deleterious characteristics of the transfusion were studied, including blood storage duration; the role of erythrocytes, leukocyte, and soluble factors; and the kinetics of the effects. RESULTS: Blood transfusion was found to be an independent and significant risk factor for cancer progression in both models, causing up to a fourfold increase in lung tumor retention and doubling mortality rates. Blood storage time was the critical determinant of these deleterious effects, regardless of whether the transfused blood was allogeneic or autogenic. Surprisingly, aged erythrocytes (9 days and older), rather than leukocytes or soluble factors, mediated the effects, which occurred in both operated and nonoperated animals. The effects of erythrocytes transfusion in the MADB106 model emerged immediately and dissipated within 24 h. CONCLUSIONS: In rats, transfusion of fresh blood is less harmful than transfusion of stored blood in the context of progressing malignancies. Further studies should address mediating mechanisms through which erythrocytes' storage duration can impact the rate of complications while treating malignant diseases and potentially other pathologies.


Subject(s)
Adenocarcinoma , Erythrocyte Aging/physiology , Erythrocyte Transfusion/adverse effects , Erythrocytes/physiology , Leukemia, Experimental , Lung Neoplasms , Mammary Neoplasms, Experimental , Adenocarcinoma/blood , Adenocarcinoma/secondary , Adenocarcinoma/therapy , Animals , Cell Line, Tumor , Disease Progression , Erythrocytes/pathology , Female , Leukemia, Experimental/blood , Leukemia, Experimental/pathology , Leukemia, Experimental/therapy , Lung Neoplasms/blood , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Male , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/secondary , Mammary Neoplasms, Experimental/therapy , Neoplasms/blood , Neoplasms/pathology , Rats , Rats, Inbred F344 , Rats, Wistar
20.
Breast Cancer Res Treat ; 107(2): 211-23, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17431763

ABSTRACT

Despite a promising potential, interleukin-12 immunotherapy has yielded limited clinical success while causing perilous toxicities. Here we study a context in which IL-12 may prove clinically beneficial--the removal of the primary tumor, when cell-mediated immunity (CMI) may eradicate minimal residual disease (MRD), but is inhibited by postoperative immunosuppression, potentially leading to enhanced malignant progression. F344 rats were preoperatively treated with IL-12 and inoculated postoperatively with syngeneic MADB106 tumor cells. An optimal regimen of eight-day sustained exposure to IL-12 was developed (1 microg/rat/day), which caused mild side effects, increased baseline resistance to experimental MADB106 metastasis, and abolished the promotion of metastasis by laparotomy and other immunosuppressive paradigms. Depletion of NK cells indicated their major role in controlling MADB106 metastasis in naïve and IL-12 treated rats. Studying NK cytotoxicity, we found that IL-12 did not potentiate activity per NK cell, nor protected it from suppression by surgery. However, IL-12 increased the numbers of NK cells in the circulation and marginating pulmonary pool of naïve and operated rats, and correspondingly increased total NK activity in these compartments. Therefore, this study indicates anti-tumor effects of IL-12 based on increased numbers of strategically located NK cells, and advocates a prophylactic approach against the potential metastasis-promoting effects of surgery.


Subject(s)
Interleukin-12/metabolism , Killer Cells, Natural/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Immune System , Immunotherapy/methods , Male , Metaproterenol/pharmacology , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...